Anda di halaman 1dari 15

Clinica Chimica Acta 363 (2006) 206 – 220

www.elsevier.com/locate/clinchim

Review

Nucleic acid-based methods for the detection of bacterial pathogens:


Present and future considerations for the clinical laboratory
Elizabeth A. Mothershed *, Anne M. Whitney
Meningitis and Special Pathogens Branch, Division of Bacterial and Mycotic Diseases, National Center for Infectious Diseases,
Centers for Disease Control and Prevention, Atlanta, GA 30333, United States

Received 3 April 2005; received in revised form 25 May 2005; accepted 26 May 2005
Available online 1 September 2005

Abstract

Background: Recent advances in nucleic acid-based methods to detect bacteria offer increased sensitivity and specificity over traditional
microbiological techniques. The potential benefit of nucleic acid-based testing to the clinical laboratory is reduced time to diagnosis, high
throughput, and accurate and reliable results.
Methods: Several PCR and hybridization tests are commercially available for specific organism detection. Furthermore, hundreds of nucleic
acid-based bacterial detection tests have been published in the literature and could be adapted for use in the clinical setting. Contamination
potential, lack of standardization or validation for some assays, complex interpretation of results, and increased cost are possible limitations
of these tests, however, and must be carefully considered before implementing them in the clinical laboratory.
Conclusions: A major area of advancement in nucleic acid-based assay development has been for specific and broad-range detection of
bacterial pathogens.
Published by Elsevier B.V.

Keywords: Molecular diagnostics; Clinical laboratory; PCR; Real-time PCR; Bacterial pathogens; NATs

Contents

1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207
2. Detection of a specific bacterial pathogen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207
2.1. Cycling amplification technologies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207
2.1.1. PCR, real-time PCR and RT-PCR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207
2.1.2. Nested PCR. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 209
2.1.3. PCR-ELISA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 209
2.1.4. Ligase chain reaction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 209
2.2. Isothermal and other amplification technologies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 210
2.2.1. Nucleic acid sequence-based amplification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 210
2.2.2. Transcription-mediated amplification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 210
2.2.3. Strand displacement amplification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 210
2.2.4. Rolling circle amplification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 210
2.2.5. Cycling probe technology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 210
2.2.6. Branch DNA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211
2.2.7. Hybrid capture . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211

* Corresponding author.
E-mail address: ekm9@cdc.gov (E.A. Mothershed).

0009-8981/$ - see front matter. Published by Elsevier B.V.


doi:10.1016/j.cccn.2005.05.050
E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220 207

3. Detection of bacterial pathogens by multiple targets or universal targets. . . . . . . . . . . . . . . . . . . . . . . . . . . . 211


3.1. Multiplex PCR. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211
3.2. Microarray . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211
3.3. Sequencing-based identification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 212
4. Detection of bacterial pathogens by nucleic acid hybridization or mass spectrometry . . . . . . . . . . . . . . . . . . . . . 213
4.1. Fluorescence in situ hybridization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 213
4.2. Peptide nucleic acid-FISH . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 213
4.3. Line probe assay. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 213
4.4. Hybridization protection assay. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 214
4.5. Mass spectrometry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 214
5. Quality control: nucleic acid extraction and contamination prevention . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215
6. How to choose which test is right for your laboratory? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215
7. Future trends for NATs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215
8. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 216
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 217

1. Introduction in culture-negative specimens. NATs for specific organ-


isms such as Mycobacterium tuberculosis, Chlamydia
Successful treatment of a patient with a bacterial trachomatis, and Neisseria gonorrhoeae [3] are available
infectious disease requires rapid and specific identification from several manufacturers, some of which are listed in
of the causative agent. Identification of bacterial pathogens Table 1. In addition, hospital infection control and
by traditional methods is still a crucial element of the epidemiology programs are benefiting from the use of
diagnostic process. However, methods such as culturing NATs for detecting antibiotic resistance genes and for sub-
and sub-culturing organisms, especially those that are typing bacteria.
fastidious, culturing clinical specimens taken after anti- While NATs offer many advantages for the clinical
biotic treatment, determining antimicrobial susceptibility of laboratory, care must be exercised when using these tests.
the organism, and biochemical testing can be laborious and Because of the increased sensitivity of NATs, contami-
time-consuming, and may prolong definitive diagnoses and nation prevention and quality control must be imple-
treatment of the patient. While there has been improvement mented. Theoretically, in a NAAT, one copy of a target
in traditional methods, such as automation of blood culture gene will be amplified; therefore, if the one copy is from
systems [1], clinical laboratories have begun to adopt a laboratory contaminant or previous experiment, a false
nucleic acid-based tests (NATs) to identify pathogens positive result will be observed. Conversely, inhibitors in
rapidly and reliably. clinical specimens or DNA degradation can lead to false
The first NAT cleared for use by the Food and Drug negative results. Another disadvantage is that NATs
Administration (FDA) was the Gen-Probe PACE test detect nucleic acids but do not indicate viability of the
(1988) that used nucleic acid hybridization to detect pathogen.
Chlamydia and gonococci (http://www.gen-probe.com/ In this review, we will discuss the identification or
prod_serv/std_pace.asp). A technological breakthrough in characterization of bacterial pathogens using NATs for a
molecular biology came in 1983 with the development of single nucleic acid target, multiple targets, or universal
polymerase chain reaction (PCR) [2]. In subsequent years, targets. Furthermore, we will review NATs that use hybrid-
nucleic acid amplification tests (NAATs), including PCR- ization technologies or mass spectrometry. Quality control
based assays, were developed to detect virtually every and contamination prevention strategies, as well as criteria
clinically relevant bacterial pathogen. (Note: going forward to consider when choosing a NAT will be discussed. Finally,
the term NATs will be used to describe all nucleic acid future trends in NATs technology and methods will be
based tests, including NAATs.) presented.
The advantages of NATs over microbiologic methods
include rapid results, low detection limits (theoretically a
single cell), and specific organism detection. In a hospital 2. Detection of a specific bacterial pathogen
setting, rapid pathogen detection is important for faster
and improved patient treatment and potentially shorter 2.1. Cycling amplification technologies
hospital stays. Community outbreaks and nosocomial
infections may be thwarted if early pathogen identification 2.1.1. PCR, real-time PCR and RT-PCR
results in patient isolation and/or directed prophylaxis of Amplification of a target sequence by conventional
patient contacts. NATs can be used to detect the presence PCR using two primers is typically detected and
of organisms directly in clinical specimens without visualized by gel electrophoresis using DNA-binding
culture, in cultures with abbreviated incubation times, or fluorescent dyes. By comparison, real-time PCR uses a
208
Table 1
Commercially available diagnostic tests for detection of common bacterial pathogens
Organism detected Test name Method/technology Time requirement Sensitivity Specificity Sample type FDA Manufacturer References

E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220
for testing (%) (%) clearance
Chlamydia trachomatis HC2 CT ID DNA/RNA Hybrid 4h 96 100 Endocervical swabs Yes Digene [26]
Capture
PACE2 CT HPA NA 78 99 Endocervical swabs Yes Gen-Probe [100]
APTIMA CT TMA/16S target NA 96 – 97 97 – 99 Urine or urethral swab No Gen-Probe [9]
Neisseria gonnorhoeae COBAS AMPLICOR PCR 4–8 h 92.4 99.5 Endocervical swabs Yes Roche [101]
BDProbeTec ETa SDA 1–2 h 99.2 99.3 Urine Yes Becton Dickinson [102]
NucliSens Basic Kitb NASBA 8h 97.9 98.7 Urethral or cervical swab No bioMerieux [12]
Mycobacterium tuberculosis Amplified MTD Direct test TMA 3.5 h 91c 98 Sputum Yes Gen-Probe [103]
BD Probe tec ET SDA 1–2 h 93 – 95 92 – 100 Respiratory or No Beckton – Dickinson [17 – 19,22,104]
non-respiratory
COBAS AMPLICORa PCR 4–8 h 97 100 Respiratory or No Roche [8]
non-respiratory
MRSA IDI-MRSA Real-time PCR 1.5 h 91.7 93.5 Nasal swabs Yes Infectio Diagnostics, Inc. [105]
Velogene-CPT Rapid MRSAd CPT 90 min 98 100 Culture Yes ID Biomedical, Inc. [24]
Group A Streptococci GAS Direct Test HPA 24 h 94.8 100 Pharyngeal swab No Gen-Probe [106]
Group B Streptococci IDI-Strep B Real-time PCR 1.5 h 94 95.9 Vaginal swabs Yes Infectio Diagnostics, Inc. [107]
Gardnerella, Trichomonas BD Affirm VPIII Microbial Hybridization 45 min 82 – 95e 98 – 100e Vaginal swabs Yes Becton Dickinson, Inc. [108,109]
vaginalis, and Candida spp. Identification Test
Escherchia coli O157:H7 BAX System PCR 8h 99 99 Water No Qualicon, Inc. [110]
HPA, hybridization probe assay; TMA, transcription mediated amplification; SDA, strand displacement amplification; NASBA, nucleic acid sequence based amplification, CPT, cycling probe technology; NA, not
available.
a
Not available in the US.
b
Primers designed by individual laboratory.
c
Sensitivity improved from 61% to 91% after retest and implementation of protocol for inhibition detection.
d
No longer available.
e
From manufacturer’s website (www.bd.com).
E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220 209

fluorescently labeled probe with two flanking primers in like the ABI 7900HT (Foster City, CA). Less expensive
the reaction. Real-time PCR offers many advantages over instruments such as the Roche LightCycler\ and Cepheid
conventional PCR such as increased sensitivity and SmartCycler\ could be options for some laboratories,
rapidity, broader dynamic range, elimination of post- however these platforms are limited by the number of
amplification handling steps, and higher throughput reactions (16 – 32) that can be tested per run. Real-time
conducive to automation [4]. Real-time PCR assays may PCR primer and probe design may be confounding since a
use the intercalating fluorescent dye, SYBR\ Green, dual- small amplicon (< 500 base pairs) is preferable. Post-
labeled probes (TaqMan\), hybridization probes (Light- amplification manipulation of the amplicon, such as
Cycler), Molecular Beacons, or Scorpionsi as the means cloning or sequencing, may also be difficult. Finally, the
of detecting amplification. Some recent publications using inability to determine amplicon size could be viewed as a
real-time PCR for bacterial detection are listed in Table 2. disadvantage by some investigators [6].
Use of a closed system greatly minimizes carry-over
contamination and most real-time PCR formats offer the 2.1.2. Nested PCR
option of melting curve analysis, which allows the Nested PCR is a conventional PCR method that amplifies
amplification product to be discriminated from nonspecific a target region of DNA with an outer primer pair in an initial
product or primer – dimers. reaction, followed by a second amplification using an
Most NATs for bacterial detection are DNA-based internal primer pair. It is useful for pathogen detection in
because the genomes of bacteria are DNA, rather than clinical specimens because of its enhanced sensitivity over a
RNA as with some viruses. However, reverse-transcriptase single amplification, but can be problematic due to carry-
PCR (RT-PCR)-based tests for bacteria have been published over contamination from the first reaction to the second [7].
such as the one for the detection of hemolysin from Vibrio Nested PCR has also been used for the detection of the 16S
parahaemolyticus described by Nakaguchi et al. [5]. RT- and 23S rRNA genes from a variety of bacteria and provides
PCR is a method for studying gene expression, and uses multiple overlapping amplicons for accurate sequencing of
RNA as its template to produce complementary DNA these genes (see Section 3.3).
(cDNA). The cDNA is then amplified by PCR. RT-PCR and
real-time PCR technologies are commonly used by research 2.1.3. PCR-ELISA
laboratories and some clinical laboratories, and many The PCR – enzyme linked immunosorbent assay (ELISA)
commercially available NATs for specific pathogen detec- format is a viable alternative to real-time PCR methods. PCR
tion utilize these methods (Table 1). Sensitivities and products are labeled (e.g., by digoxigenin) during amplifi-
specificities of these tests, however, may be affected by cation and a capture probe specific to the PCR amplicon is
the specimen type used, nucleic acid extraction method, and used to immobilize the amplicon to a well of a microtiter
quality of the primers and fluorescent probes used in the plate. An enzyme-linked antibody targeting the label (e.g.,
tests. anti-digoxigenin) is then used to quantitate PCR products.
Though real-time PCR is in widespread use because of As Yam et al. demonstrate, a biotinylated PCR-ELISA for
its enhanced sensitivity, the overriding limitation of this direct detection of M. tuberculosis using a single-tube nested
technology is the high cost of special reagents and PCR method provides a simple, accurate, high throughput
instrumentation. The price of a single real-time PCR test with sensitivity and specificity comparable to the
reaction including DNA extraction can cost up to $10, commercial, PCR-based COBAS AMPLICOR system
while conventional PCR costs less than $3 per reaction. (Roche Diagnostics; Table 1) at around one-fourth the cost
Instrumentation costs range from $24,000 for the Strata- [8].
gene MX3000 (La Jolla, CA), for example, to over
$130,000 for an automated, high-throughput instrument 2.1.4. Ligase chain reaction
Ligase chain reaction (LCR) is a DNA amplification
Table 2 technique, but differs from PCR because it does not produce
Recently published real-time PCR assays for detecting bacterial pathogens amplicon through polymerization of nucleotides. In LCR, a
Organism Platform Detection Reference primer is synthesized in 2 fragments and annealed to the
Leptospira spp. LightCycler\
SYBR Green I [111] template. The ligase enzyme will join the 2 fragments only if
Neisseria meningitidis TaqMan\ Dual-labeled probe [112] they match exactly to the template sequence. Subsequent
Burkholderia TaqMan\ Dual-labeled probe [113] PCR reactions will amplify the template only if the primer
pseumomallei fragments are joined. A once-popular commercial test, the
Bordatella pertussis LightCycler\ Hybridization probes [114]
LCx (Abbott Laboratories, Abbott Park, IL) for Chlamydia
Bordetella parapertussis LightCycler\ Hybridization probes [114]
Escherichia coli LightCycler\ Hybridization probes [115] detection, was apparently taken off the market in 2003 due to
Methicillin-resistant SmartCycler\ Molecular Beacons [105] problems with negative controls (http://www.chlamydiae.
Staphylococcus aureus com/restricted/docs/labtests/diag_lcr.asp), but its perform-
Group B Streptococcus SmartCycler\ Molecular Beacons [107] ance has been compared to other commercial tests in the
Clostridium difficile SmartCycler\ Molecular Beacons [116]
literature [9].
210 E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220

2.2. Isothermal and other amplification technologies slightly less sensitive to inhibitors than the AMTDII [17].
Gamboa et al. found that, compared to culture and staining,
2.2.1. Nucleic acid sequence-based amplification the AMTDII test was 95% and 100% sensitive and specific,
Nucleic acid sequence-based amplification (NASBA) is respectively, using respiratory specimens, and slightly less
an isothermal, transcription-based amplification method sensitive using non-respiratory specimens [18]. Kerleguer et
which amplifies RNA from either an RNA or DNA target al. found the MTD test to be 93% sensitive and 100%
and utilizes avian myeloblastosis virus reverse transcrip- specific using lymph node aspirates, while culture was 89%
tase, RNase H and T7 RNA polymerase. NASBA has been sensitive [19].
used to detect various bacterial pathogens including
Escherichia coli and Mycoplasma pneumoniae [10,11]. 2.2.3. Strand displacement amplification
NASBA has demonstrated equivalent or improved sensi- Strand displacement amplification (SDA), first
tivity to PCR-based methods and has the potential described in 1992 [20], is an isothermic amplification
advantage of being easier to optimize than conventional method in which a primer containing a restriction site is
PCR [12]. Rodriguez-Lazaro et al. describe a NASBA annealed to the DNA template. Next, amplification
assay for detecting the dnaA gene of Mycobacterium primers are annealed to 5V adjacent sequences (forming a
avium subsp. paratuberculosis from water and milk and nick) to begin amplification. Newly synthesized DNA is
detail the inclusion of an internal amplification control nicked by the corresponding restriction enzyme and the
specific for the NASBA method [13,14]. Diagnostic tests polymerase starts amplification again, displacing the newly
using NASBA are also commercially available for the synthesized strands. In a single reaction, 109 copies of
pathogens C. trachomatis and N. gonorrhoeae (Table 1). target DNA can be produced. SDA is the basis for some
BioMerieux (Durham, NC) has combined NASBA and commercial detection tests such as BDProbeTec (Becton
Molecular Beacons into a test system called EasyQ for Dickinson, Franklin Lakes, NJ) and has been evaluated
simultaneous amplification and fluorescent detection of recently for the identification of M. tuberculosis directly
specific organisms (http://www.biomerieux-usa.com/clinical/ from clinical specimens [21]. A study of BDProbeTec’s
nucleicacid/easyq/easyq_technology.htm). assay for C. trachomatis and N. gonorrhoeae on vaginal
swab specimens showed sensitivity and specificity equiv-
2.2.2. Transcription-mediated amplification alent to those of PCR for the detection of C. trachomatis
Transcription-mediated amplification (TMA) is another and superior to those of culture for the detection of N.
isothermal amplification method that can be used to target gonorrhoeae [22].
either DNA or RNA. TMA uses RNA transcription (RNA
polymerase) and DNA synthesis (reverse transcriptase) to 2.2.4. Rolling circle amplification
produce an RNA amplicon from a target nucleic acid. Since In rolling circle amplification (RCA), a single forward
RNA is more labile than DNA in the laboratory environ- primer is extended by DNA polymerase along a circular
ment, this feature diminishes the possibility of carry-over template for many rounds, displacing upstream sequences
contamination. TMA produces 100– 1000 copies per cycle and producing a long single-stranded DNA of multiple
as compared to PCR and LCR that produce only 2 copies repeats. The linear RCA reaction can run for several hours
per cycle. This results in a 10 billion-fold increase of copies or days, producing millions of copies of the small circle
within about 15 – 30 min. TMA has gained popularity in the sequence. In exponential RCA, a primer pair is used. The
clinical laboratory with the development of commercial tests second primer targets the single-stranded DNA product of
including the APTIMA tests for the detection of C. the first primer and initiates hyper-branching in the DNA
trachomatis and N. gonnorhoeae (Table 1) [15]. replication, creating as many as 1012 copies/h. As is the
In a comparison of Gen-Probe APTIMA CT and GC case for all isothermal technologies, there is no need for
assays with the APTIMA combo 2 assay, the Abbott LCx special instrumentation, since temperature cycling is not
assay, direct fluorescent antibody test, and culture for the required. A major advantage of RCA is that, unlike PCR,
detection of C. trachomatis and N. gonorrhoeae, Boy- this technology is resistant to contamination and, unlike
adzhyan et al. reported that the APTIMA tests detected some other isothermal technologies, requires little or no
more confirmed positive specimens than culture, direct assay optimization. A recent review discusses details of the
fluorescent antibody test, or LCx [9]. A similar study found technology and the latest progress in RCA diagnostics
the Abbott LCx, BD ProbeTec ET and the Gen-Probe [23].
APTIMA Combo 2 to be very sensitive (96%, 96% and
100%, respectively) and specific (99%, 100% and 99%, 2.2.5. Cycling probe technology
respectively) for detection of C. trachomatis in urine [16]. Cycling probe technology (CPT) is an isothermal probe
Another Gen-Probe product that incorporates TMA is the amplification system for detection of target DNA that
amplified M. tuberculosis direct test (MTD). Lemaitre et al. utilizes a RNA – DNA chimeric probe (RNA sequence
compared real-time PCR to the AMTDII test (Gen-Probe, flanked by two DNA sequences) to hybridize to a specific
San Diego, CA) and found that real-time PCR was only region of an amplified gene. Once hybridized, the internal
E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220 211

RNA part of the probe is cleaved by RNase H, which 3.1. Multiplex PCR
recognizes specifically the RNA –DNA duplex. Once the
probe is cleaved, the reporter dye and quencher dye on each Multiplex PCR utilizes more than one set of primers in a
side of the probe are separated and fluorescence is emitted. reaction and can be used for the simultaneous detection of
The fluorescence signal increases proportionally as the multiple bacterial pathogens. Either conventional or real-
probe is cleaved, allowing for measurement of the amplified time PCR can be used for a multiplex reaction. The Hyplex
product. The probe-based Velogene Rapid MRSA Identi- Blood Screen multiplex PCR-ELISA system (BAG, Lich,
fication Assay (ID Biomedical Corp., Vancouver, British Germany) is a commercially available test used to screen
Columbia, Canada) has been used previously for detection positive blood cultures for the most common bacterial
of methicillin-resistant Staphylococcus aureus (MRSA), but pathogens that cause sepsis. This test system uses PCR to
is no longer available in the US [24]. amplify organism-specific housekeeping genes and detects
the amplification products by hybridization to oligonucleo-
2.2.6. Branched DNA tide probes in an ELISA format [28]. Also, multiplex PCR
Another technique that utilizes signal rather than target in combination with colorimetric Covalink NH microwell
amplification is called branched DNA (bDNA). In bDNA, plate (DNA – DNA sandwich hybridization) detection is a
many branched, labeled DNA probes generate signal via method used in clinical settings because of its low cost and
alkaline phosphatase upon binding to a specific nucleic acid ease of use. As Lee et al. show by simultaneously detecting
target. bDNA has been used to detect the mecA gene in Vibrio spp. and Salmonella enterica in shellfish, traditional
MRSA [25], though it is more commonly used for multiplex PCR with multiple primer sets can also demon-
determination of viral load. strate a high level of sensitivity and specificity [29].
Multiplex PCR is also useful for detecting a species-specific
2.2.7. Hybrid capture target and a serogroup-specific target in a single reaction.
Signal amplification is also the basis for some For example, Taha has developed a multiplex PCR for the
commercial tests such as the Hybrid Capture (HC2) detection of Neisseria meningitidis in which 6 pairs of
assays from Digene (Gaithersburg, MD) (Table 1). In the serogroup-specific primers are added together in a single
Hybrid Capture tests, a lysis solution is used to release reaction [30].
DNA from the bacteria, and specific RNA probes combine The Luminex system (Luminex Corp., Austin, TX)
with the target DNA to form a hybrid. The RNA/DNA uses a recently developed bead-based technology, xMAP,
hybrid is then captured by specific antibodies to a solid which has the potential for routine testing in the clinical
support, and the hybrids are detected by a second antibody laboratory. Introduced as an immunoassay, this technology
conjugated to alkaline phosphatase which cleaves a has gained popularity in a nucleic acid-based format
chemiluminescent substrate to release light. Because many because of the capability to detect many targets in a
antibody molecules attach to each hybrid, the signal is single test (multiplex), while still maintaining a high
amplified. Hybrid Capture systems are available to detect level of sensitivity and specificity [31]. Polystyrene
C. trachomatis, N. gonorrhoeae, human papillomavirus, microbeads are imbued with two fluorescent dyes. The
cytomegalovirus and hepatitis B. A benefit to using a ratio of one dye to another creates 100 unique spectral
signal amplification assay versus a target amplification signatures. These bead sets can be coated with peptides,
assay (e.g. PCR) is that amplicons are not produced in the receptors, oligonucleotides or antibodies. In a recent
laboratory; therefore, the chance of cross-contamination of study, Dunbar et al. demonstrated simultaneous detection
subsequent reactions is reduced. Apparently, sensitivity is of multiple food-borne pathogens, E. coli, Salmonella,
not lost with signal amplification assays. Independent Listeria monocytogenes and Campylobacter jejuni with
studies have found that the Digene Hybrid Capture II kit rapid (40 min), specific identification; however, sensitivity
was a highly sensitive and specific assay for detection of was not ideal, because the test required 106 to 107
C. trachomatis [26, 27]. genome copies for detection [32]. As with any method,
though, multiplex PCR is not without disadvantages.
Problems such as low sensitivity [33], cross-reactivity,
3. Detection of bacterial pathogens by multiple targets or and preferential binding of SYBR\ Green I to longer,
universal targets higher G + C% amplification products [34] have been
reported for multiplex reactions.
The overriding disadvantage of organism-specific assays
is that they are limited in scope and useful only when a 3.2. Microarray
particular agent is suspected. For this reason, technologies
such as multiplex PCR, microarray, and broad-range PCR Microarray refers to a small, two-dimensional high-
assays have been developed for the purpose of testing density matrix of DNA fragments which are printed or
simultaneously for more than one organism or as a means to synthesized on a glass or silicon slide (chip) in a specific
screen clinical specimens for bacterial etiologic agents. order. Hybridization of the DNA fragments to fluorescently
212 E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220

labeled probes is detected by advanced instrumentation and Table 3


software. However, microarrays for bacterial diagnostics Bacterial pathogens identified or characterized by 16S rRNA gene
sequencing
have not been incorporated into routine testing in clinical
Organism Syndrome or Specimen/site Reference
laboratories because of the complex technical aspects for
disease of isolation
successful experiments such as uniform stringency over the
Atopobium vaginae Bacterial vaginosis Vaginal lavage [117]
array chip, data analysis and interpretation, and high cost.
Bacillus anthracis Respiratory and Various sitesa [45]
Even with such confounders, some manufacturers offer ‘‘off cutaneous anthrax
the shelf’’ assays which are reported to be sensitive, specific, Bacillus cereus Pneumonia Sputum, blood [118]
and easy-to-interpret. Affymetrix has developed the Brucella spp. Brucellosis Various sitesb [49]
Advanced Array Technology (AAT, Eppendorf Array Tech- Burkholderia Melioidosis Various sitesc [50]
pseudomallei
nologies, Belgium) Staphychip which detects 15 species of
Clostridium hathewayi d Septic shock Blood [119]
Staphylococcus, including methicillin-resistant S. aureus Corynebacterium Sinus disease Sinus secretione [120]
(MRSA) (www.aat-array.com/staphy.htm) [35] and the fastidiosum
Mycochip which detects 14 mycobacterial species. Qiagen, Haemophilus segnis Pyelonephritis Blood [121]
Inc. (Valencia, CA) offers arrays for many bacterial patho- Helicobacter pylori Dyspepsia Supragingival [122]
plaque
gens including Bacillus anthracis, C. jejuni, E. coli,
Neisseria meningitidis Meningitis csf [40,53]
Haemophilus influenzae, Neisseria spp., and Salmonella Staphylococcus Sinus disease Sinus secretione [120]
spp. The Nanochip (Nanogen, Inc., San Diego, CA) is one epidermis
example of a self-contained electronic microarray chip, Staphylococcus Onycholysis, Wound discharge [123]
primarily used for single nucleotide polymorphism (SNP) intermedius localized cellulitis
Streptococcus Thoracic empyema Pleural fluid [124]
detection in human genetic applications, but has recently
pneumoniae
been evaluated for bacterial detection. Designed to target long Streptococcus pyogenes Thoracic empyema Pleural fluid [124]
(350 base pair) and short (200 base pair) regions of the 16S a
Tissue, pleural fluid, blood, lymph node.
rRNA gene of 8 marine bacteria, the Nanochip demonstrated b
Tissue, blood.
variable specificity, and optimization was recommended c
Blood, sputum, abscess.
d
[36]. If a commercial microarray is not desirable, custom Typically found in feces of healthy individuals, this organism was
microbial diagnostic microarrays are available from or- associated with acute gangrenous appendicitis and septic shock in this case.
e
Serous, mucous, or pus.
ganizations like the Department of Bioresources at Seibers-
dorf, Austria and others. For these custom products,
oligonucleotide probes based on 16S rRNA, 16S – 23S pathogenic bacteria from clinical specimens that have been
inter-gene spacer, or other functional genes are designed identified by 16S rRNA sequencing.
(http://www.diagnostic-arrays.com/coop.htm). Non-com- Bacterial identification by 16S rRNA gene sequencing
mercial arrays for bacterial detection have also been has become increasingly popular with clinical laboratories
developed such as that described by Roth et al. Using broad since costs have decreased and improved throughput has
range primers targeting topoisomerases, a diagnostic array become available in the last few years [39]. One recent
was developed for the identification of 9 upper respiratory study even indicates that the cost for 16S rRNA gene
bacterial pathogens [37]. A clinical laboratory might also sequencing can be as low as a third the cost of con-
consider using a validated microarray for detection of ventional identification methods [46]. To minimize costs,
antimicrobial resistance genes. Recently, Yu et al. developed packages such as the MicroSeq 500 and the MicroSeq
and validated a diagnostic microarray for the detection of 16S rRNA gene systems (Applied Biosystems, Foster
fluoroquinolone-resistant E. coli clinical isolates [38]. City, CA) are commercially available [47,48]. For those
clinical laboratories that do not have sequencing capa-
3.3. Sequencing-based identification bilities, 16S rRNA gene sequencing services can be
outsourced.
If an assay for a specific bacterial pathogen is not The speed with which identification can be made by 16S
available or when multiple agents may be implicated as the rRNA gene sequencing compared to traditional biochemical
cause of disease, a broad-range detection approach can be methods is also an advantage [39,46,49,50] especially in
useful. Universal targets such as the 16S rRNA genes or the cases where conventional methods were inadequate [40].
16S – 23S rRNA gene interspacer region have been used For example, fastidious organisms, such as Mycobacteria
extensively for bacterial identification, especially if bacteria spp., can be identified in 24 h by 16S rRNA gene
are difficult to isolate by conventional methods [39 – 41]. sequencing [51]. In fact, for some pathogens, 16S rRNA
16S rRNA gene sequencing has also been used successfully gene sequencing may be the only rapid NAT available as
to detect pathogens from culture-negative specimens [42]. PCR-based tests have not yet been developed [46]. As with
Furthermore, recent work has indicated that 16S rRNA gene any diagnostic method, there are some pitfalls of using a
sequences may form the basis for sub-typing schemes for universal gene target such as 16S rRNA. Some of the
some pathogens [43 – 45]. Table 3 lists some of the sequences in the public databases (i.e. GenBank) are known
E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220 213

to contain errors [52], especially those submitted prior to the spp. [62], Brachyspira spp. [63], Enterococcus spp.,
development of high-fidelity, automated sequencing sys- Pseudomonas aeruginosa [64], Helicobacter spp.[64],
tems [49,50,52]. Because 16S rRNA gene primers target Streptococcus spp. [65,66] Staphylococcus spp., Yersinia
conserved sequences and the 16S rRNA gene is ubiquitous spp., and others [67]. Many of these studies were aimed at
among all bacteria, a sequence may be amplified from a identifying bacteria in positive blood culture bottles obviat-
contaminating bacterium instead of the etiologic pathogen ing the need for subculture. A few FISH NATs are
[53]. Other universal targets such as heat-shock proteins, commercially available including those from Microscreen
like hsp65 [54] or cold-shock proteins [55] have also been Ribotechnologies (Groningen, The Netherlands) for the
used to identify bacteria from clinical specimens. detection of human intestinal bacteria: Bifidobacterium
Pyrosequencing (Pyrosequencing AB, Uppsala, Sweden) spp., E. coli, Lactobacillus spp., Streptococcus spp. and
is a technology whereby a single-stranded DNA template is Clostridia spp.
prepared, a sequencing primer is hybridized to a compli-
mentary sequence on the template, and enzymes catalyze a 4.2. Peptide nucleic acid-FISH
light reaction when each nucleotide is incorporated into the
growing DNA strand [56]. Pyrosequencing has been used to In 1991, Nielson et al. discovered a DNA analogue called
identify and characterize bacterial pathogens such as peptide nucleic acid (PNA) [68]. Fluorescently labeled
Helicobacter pylori [57], N. meningitidis [58], and N. PNAs have been successfully used as hybridization probes
gonorrhoeae [59], and rapidly discriminate pathogenic from in FISH assays. PNA probes have distinct advantages over
non-pathogenic bacteria in complex specimens [60]. The DNA probes including the stability of the PNA/RNA hybrid
major advantage of this technology is that the price per due to the uncharged PNA [69]. Also, PNAs enter a
sample reaction may cost 10-fold less than fluorescent- bacterial cell more easily because of their relative hydro-
based sequencing [61]. phobicity. PNAs also have higher specificity than DNA
oligomers due to the higher Tm of the PNA probe compared
to its DNA counterpart [70].
4. Detection of bacterial pathogens by nucleic acid AdvanDX (Woburn, MA) offers three PNA-FISH assays
hybridization or mass spectrometry for diagnostic use and all have gained FDA clearance.
AdvanDx PNA-FISH NATs for definitive identification of
The discovery of PCR has revolutionized molecular S. aureus, Candida albicans and Enterococcus faecalis
diagnostics over the past decade, and it seems that the directly from positive blood culture bottles are currently in
number of PCR-based NATs being developed mimics the use in some US medical centers [70 –73]. In an independent
exponential amplification of target molecules! However, evaluation of the AdvanDX S. aureus PNA-FISH test,
non-amplification methods for detecting bacteria are also Gonzalez et al. found that the test was 100% sensitive, 99%
commercially available or are evolving to the point that they specific, and gave 99% and 100%, positive and negative
can be easily performed in a clinical laboratory. Many of the predicative values, respectively, compared to three con-
commercially available non-amplification NATs rely on firmatory tests [74]. For the detection of S. aureus, Chapin
detection of a specific target by chemiluminescence, and Musgnug performed a comparison of PNA-FISH
colorimetric, or fluorescent signals. (AdvanDX), API RAPIDEC Staph System (bioMerieux),
and direct tube coagulase test, using the AccuProbe S.
4.1. Fluorescence in situ hybridization aureus Culture Identification kit (Gen-Probe) as the stand-
ard. PNA-FISH demonstrated 99% sensitivity and 100%
Fluorescence in situ hybridization (FISH) assays use specificity [75].
fluorescently labeled 16S rRNA or 23S rRNA probes and In addition to PNA-FISH, theoretically, PNAs could be
fluorescent microscopy to detect intact bacteria directly in substituted for DNA oligonucleotides to enhance an assay’s
clinical specimens, such as blood or tissue, or after performance. PNAs have been incorporated into chemilu-
enrichment culture. FISH is a useful method for detection minescent in situ hybridization assays (CISH) [76,77],
of fastidious organisms such as Bartonella spp. and microarrays [78], biosensors [79,80], PCR clamping assays
Yersinia pestis. In fact, multiple species can be detected for mutant allele detection [81], Molecular Beacons [82] and
simultaneously using two or more specific probes labeled light-up probes for real time PCR [83].
with unique fluorescent dyes. The procedure takes
between 1 and 2 h and consists of fixing the specimen, 4.3. Line probe assay
preparing a smear or section on a microscope slide,
permeabilizing the cells, hybridizing the target sample The line probe assay (LiPA) consists of a nitrocellulose
with the probe and detecting hybridization by fluorescence strip with specific oligonucleotide probes attached as
microscopy. discreet parallel lines along the strip. Hybridization results
Family-, genus-, and species-specific FISH probes have in a color change that can be detected visually or by an
been developed and published for detection of Chlamydia automated reader. Innogenetics (Gent, Belgium) produces
214 E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220

several line probe NATs for bacterial detection including value, and >95% negative predictive value. The S. aureus
ones for M. tuberculosis complex and Mycobacterium spp., test, however, demonstrated lower sensitivity (81%) [88].
rpoB gene mutations conferring rifampicin resistance, and
Treponema pallidum antibodies. The INNO-LiPA Rif.TB 4.5. Mass spectrometry
test detects the M. tuberculosis complex, specifically five
genotypes corresponding to sensitivity to rifampicin and Mass spectrometry (MS) causes ionization and disinte-
four resistant genotypes. Test results were 100% concordant gration of a target molecule by bombarding it with electrons.
with antibiogram results [84], and in a separate study, the The mass/charge ratio of the resulting molecular fragments is
sensitivity and specificity of this test were 100% and 92%, then analyzed to produce a molecular signature. MS has
respectively [85]. The INNO-LiPA MYCOBACTERIA v2 often been used to identify bacteria by protein signature but
differentiates 16 mycobacterial species using probes specific was not considered a useful tool for DNA studies. However,
for the 16S – 23S rRNA spacer region. The first version of in the past 15 years the difficulties of analyzing DNA have
this test was evaluated in 2000 and correctly identified 50 of been largely overcome [89], and the use of MS for nucleic
53 isolates to the species level [86]. The test was recently acid analysis has developed rapidly. Matrix-assisted laser
improved and evaluated with 642 Mycobacterium spp. desorption/ionization time of flight (MALDI-TOF) MS
isolates and 27 non-mycobacterial isolates [87] and dem- generates sample data in seconds. Because of its rapidity
onstrated 100% sensitivity and specificity and 99.2% and the capability of analyzing thousands of samples per day,
accuracy [87]. many researchers are now investigating the use of MS as a
diagnostic tool for bacterial detection. MALDI-TOF MS has
4.4. Hybridization protection assay proven to be valuable for SNP detection in human DNA, and
was recently used to differentiate virulent N. meningitidis
Hybridization protection assays (HPA) utilize a chemilu- clones [90]. An SNP within the fumC gene was used to
minescent acridinium ester detector molecule on a DNA discriminate between the hypervirulent ET-15 strain and
probe that targets the specific bacterial rRNA. The RNA/ other ET-37 complex strains, and MALDI-TOF proved to be
DNA hybrid is detected in a luminometer. AccuProbe (Gen- an efficient alternative to traditional DNA sequencing [90].
Probe, San Diego, CA) HPA tests are available for the Analysis of uncultured bacteria is also possible by MALDI-
detection of M. avium, M. avium complex, M. intracellulare, TOF MS. By incorporating dUTP rather than dTTP during
M. gordonae, M. kansasii, M. tuberculosis complex, 16S rRNA gene PCR, researchers were able to detect single
Campylobacter spp., Enterococcus spp., Group A Strepto- nucleotide differences in fragment patterns after uracil –
coccus (S. pyogenes), Group B Streptococcus (S. agalactia), DNA –glycosylase treatment [91]. Lefmann et al. were able
H. influenzae, N. gonorrhoeae, S. aureus, S. pneumoniae and to genotype 12 type strains and 24 clinical isolates of
L. monocytogenes. After investigators adjusted the cutoffs for Mycobacteria using RNA transcripts of 16S rRNA genes
a positive result to increase the sensitivity of the S. aureus, S. [92]. The data published in the last two years on bacterial
pneumoniae, Enterococcus spp., Group A Streptococcus DNA analysis, and the TIGER project (see Section 7) may
and Group B Streptococcus tests, four had > 90% compel more research in MALDI-TOF MS for clinical
sensitivity, >98% specificity, > 94% positive predictive diagnostics.

Table 4
Instruments for automated nucleic acid extraction
Manufacturer Instrument Model Capture method Maximum Maximum Elution Processing time for Approximate
no. of samples sample volume volume maximum samples cost
Applied Biosystems ABI Prism 6100 Silica/filtration 96 700 Al 150 Al 0.5 h $150,000
6700 Silica/filtration 96 700 Al 40 – 200 Al 1.5 h $154,500
Autogen AutoGenPrep 245 Centrifugation 24 Tissue Pellet 3.5 h Inquirea
AutoGenPrep 965 Centrifugation 384 200 Al Pellet 4–6 h Inquire
AutoGenFlex 3000 Centrifugation 40 5 mL Pellet 5h Inquire
bioMerieux NucliSens Extractor Silica/filtration 10 2 mL 35 Al 45 min $80,000
Mini-Mag Silica/magnetic 12 1 mL 25 Al <1 h $12,000
Qiagen BioRobot EZ-1 Silica/magnetic 6 350 Al 200 Al 0.33 h $29,000
M48 Silica/magnetic 48 350 Al 100 – 400 Al 3.5 h $73,450
M96 Silica/magnetic 96 50 Al 50 – 100 Al 3.5 h $93,150
MDx Silica/filtration 96 200 Al 200 Al 2.5 – 4 h $170,000
9604 Silica/filtration 96 200 Al 400 Al 2h $70,000
Roche MagNA Pure LC Magnetic glass particles 32 150 Al 100 Al 1 – 1.5 h $84,500
Compact Magnetic glass particles 8 1000 Al 50 – 200 Al 35 min $35,000
a
www.autogen.com.
E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220 215

5. Quality control: nucleic acid extraction and Table 6


contamination prevention Criteria to consider when choosing a nucleic acid-based test
Criterium Test or method to consider
When choosing to use a NAT, it is important to recall the High-throughput Real-time PCR (96 or 384 well assays);
adage ‘‘Garbage in, garbage out’’. In other words, results multiplex real-time PCR; automation
Low limit of detection NAATs
produced by the NAT are only as good as the quality of the
Multiple target detection Multiplex PCR; multiplex real-time
nucleic acid that goes into the test. The presence of PCR; microarray
inhibitors in clinical specimens, for example, may interfere Broad-based bacterial detection 16S rRNA or 23S rRNA gene
with PCR or hybridization reactions and cause false sequencing
negative results [93]. Therefore, selection of the nucleic Antimicrobial resistance Real-time PCR; microarray;
detection commercial NATs
acid extraction method is of critical importance. Many
Ease of use Commercial NATs
commercially available extraction kits incorporate a buffer
to lyse the bacteria and a silica matrix membrane (typically
in column format) to trap the DNA or RNA. Several wash traditional methods adequate to produce timely, reliable
steps are required to remove protein and other macro- results? While many factors must be considered before
molecules, and the purified DNA and RNA is then eluted implementing NATs, including cost assessment, some
from the membrane. Many of the manual extraction important points to consider as listed in Table 6.
methods require several centrifugation steps. To reduce
hands-on time, operator error, and sample contamination,
semi-automated DNA or RNA extraction kits and equip- 7. Future trends for NATs
ment have been designed and are commercially available
(Table 4). Certainly, a rapid, simple test that provides an accurate
Strategies to prevent contamination should also be diagnosis is the ‘‘holy grail’’ of any clinical laboratory or
considered when using NATs, as the inherent sensitivity of clinician. While technology may not yet allow for such a
these assays makes detection of undesirable nucleic acid a reality, it is intriguing to consider the advances being made
potential problem [94,95]. Some simple contamination in molecular diagnostics and perhaps glimpse that such a
control methods are listed in Table 5 and were recently goal is not far out of reach. Some innovative technologies
reviewed in detail by Aslanzadeh [96] and Borst et al. [95]. and future applications that are currently in development
Importantly, it should be noted that each assay mentioned and evaluation are shown in Table 7.
in this review may have deficiencies such as suboptimal Notably, miniature devices, known as biosensors, are
sensitivity, specificity or efficiency. It is not the intent of this being developed for chemical and microorganism detec-
review to recommend one assay or instrument over another. tion. In general terms, a biosensor is an instrument that
It is the responsibility of the clinical laboratory to conduct detects a target (analyte) by capturing the target (hybrid-
its own validation of any assay or procedure before ization or binding to antibody) and transducing the capture
implementing it for routine diagnostic use [97,98]. event into a detectable signal. Many of the sensors in
development use DNA or protein as the sensor and an
optical, acoustic, surface plasmon resonance (SPR), micro-
6. How to choose which test is right for your laboratory? gravimetric, chemiluminescent, fluorescent, or electro-
chemical signal output (Table 7). These devices have
With the abundance of tests and technologies available, a been of keen scientific interest for detection of bacteria
laboratory must consider what factors are most important for because they have the potential to be fast, reliable,
achieving the desired results before choosing a NAT. The sensitive, and mass-produced. In particular, biosensors
first question should be does a need exist for NATs or are have been a priority for blood collection centers to detect

Table 5
Quality control strategies to consider when performing nucleic acid-based tests
Strategy How it works
Designate a ‘‘clean’’ area for reaction set-up Room under negative air pressure; positive-displacement pipettes; aerosol-block pipette tips; UV-equipped
PCR cabinet
Use personal protective equipment (PPE) Disposable gloves and lab coats should be worn to prevent introduction of contaminating DNAs or
nucleases
Use uracil-N-glycosylase (UNG) in real-time Real-time PCR incorporates dUTP not dTTP during amplification; UNG cleaves DNA containing UTP,
PCR reactions thus destroying contaminating carry-over amplicons
Use a ‘‘hot-start’’ method Minimizes false priming events by withholding a crucial reaction component until appropriate temperature
is reached
Use controls in every test External positive and negative controls monitor reaction performance and contamination; internal controls
monitor presence of inhibitors
216 E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220

Table 7
Innovative technologies for bacterial pathogen detection
Device Manufacturer Detection method URL or reference
RAPID, Ruggedized Advanced Pathogen Idaho Technology, Salt Lake City, UT Real-time PCR http://www.idahotech.com/
Identification Device
RAZOR Idaho Technology, Salt Lake City, UT Real-time PCR http://www.idahotech.com/
HANAA, Handheld Advanced Nucleic Lawrence Livermore National Laboratory, Real-time PCR [125]
Acid Analyzer Livermore, CA
Bio-Seeq Smiths Detection, Pinebrook, NJ Real-time PCR [126]
DiversiLab Bacterial Barcodes, Inc., Houston, TX rep-PCR [127 – 129]
LIAT Lab-in-a-tube Iquum, Inc., Allston, MA Various http://www.iquum.com
GeneXpert Cepheid, Sunnyvale, CA Real-time PCR http://www.cepheid.com
Unnamed (microfluidic) HandyLab, Ann Arbor, MI Various http://www.handylab.com
Nanochip Nanogen, San Diego, CA Various http://www.nanogen.com
Quantum Dot Quantum Dot, Hayward, CA Nanocrystals [130]
Verigene Nanosphere, Northbrook, IL Gold nanoparticles http://www.nanosphere-inc.com
Scansystem Hemosystem, Marseilles, France Chemiluminescent [131,132]
Biacore Biacore, Piscataway, NJ Optical [133 – 135]
Spreeta Texas Instruments, Attleboro, MA Optical [133 – 135]
Tiger Isis Pharmaceuticals Carlsbad, CA and Science
Applications International
Corporation, San Diego, CA Mass spectrometry [136]
Unnamed Not commercially available Magnetic field [137]
Unnamed Not commercially available Microbalance [138,139]
Unnamed Not commercially available Acoustic [140]
Unnamed Not commercially available Electrochemical [141,142]
Unnamed Not commercially available Amperometric [143]
Unnamed Not commercially available Optical [144]
Unnamed Not commercially available NASBA [145]

blood product contamination and for national security true positive, but what if the result from that patient’s
agencies to detect biowarfare agents in air, food and water. specimen yielded Burkholderia picketti? Researchers have
Many of these instruments could potentially become part of shown that DNA from microorganisms can be amplified
a clinical laboratory or even used as portable devices for from plastic ware, PCR reagents, the lab environment, and
bedside diagnostic use. water (reviewed in [95]). Nikkari et al. amplified bacterial
DNA from healthy individuals’ blood [99].
Negative results will also pose a challenge for the
8. Conclusions clinician. When interpreting a negative result, consideration
must be given to the specimen type, antibiotic treatment of
There is great potential of molecular assays to increase the patient, and bacterial concentration in the small sample
the speed and accuracy of bacterial identification in the volume used for the NAT. Therefore, all molecular and
clinical laboratory. The limitations of NATs must be immunological diagnostic results must be reviewed and
considered, however, when deciding whether to incorporate critically interpreted based on the patient’s clinical presen-
these tests in the diagnostic algorithm. False positive and tation before definitive diagnoses and treatment decisions
false negative results may have a significant impact on are made. It is not the intention of this review to recommend
patient management [95]. To reduce such errors, contami- using NATs as first line diagnostic tools, but rather to
nation must be minimized, technicians must have proper emphasize the important role of these tests in assembling an
training, and quality control procedures must be incorpo- accurate and timely diagnosis of bacterial disease.
rated into routine laboratory workflow. While the cost of Molecular diagnostic methods will most likely continue
some molecular diagnostic instruments is high, the benefits to be incorporated into clinical laboratory work as more
of faster turnaround time, high throughput, and enhanced commercialized assays are developed and cleared through
sensitivity over traditional methods may override this FDA, prices decrease due to evolving technology, and
obstacle. Another important consideration is that laboratory clinical microbiologists and clinicians become accustomed
space must be allocated for instruments and dedicated as to the advantages of NATs. The limitations of these
DNA-free areas. systems will continue to be solved. Integrated, closed
It is purported often that the new molecular methods can microsystems will help in contamination prevention, and
be sensitive and specific, but what does a ‘‘positive’’ test assays will become more standardized and easier to
result mean clinically? Caution must be taken when perform. With time, integrated NATs for bacterial detection
interpreting NATs results. A positive result for Bartonella may be developed for hospital bedside, doctors’ offices or
in a patient with culture-negative endocarditis is a possible even home use.
E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220 217

References tuberculosis in respiratory and nonrespiratory specimens. J Clin


Microbiol 1998;36:684 – 9.
[19] Kerleguer A, Fabre M, Bernatas JJ, et al. Clinical evaluation of the
[1] Kaltsas P, Want S, Cohen J. Development of a time-to-positivity Gen-Probe amplified Mycobacterium tuberculosis direct test for
assay as a tool in the antibiotic management of septic patients. Clin rapid diagnosis of tuberculosis lymphadenitis. J Clin Microbiol
Microbiol Infect 2005;11:109 – 14. 2004;42:5921 – 2.
[2] Saiki RK, Scharf S, Faloona F, et al. Enzymatic amplification of beta- [20] Walker GT, Fraiser MS, Schram JL, et al. Strand displacement
globin genomic sequences and restriction site analysis for diagnosis amplification—an isothermal, in vitro DNA amplification technique.
of sickle cell anemia. Science 1985;230:1350 – 4. Nucleic Acids Res 1992;20:1691 – 6.
[3] Paillard F, Hill CS. Direct nucleic acid diagnostic tests for bacterial [21] McHugh TD, Pope CF, Ling CL, et al. Prospective evaluation of
infectious diseases: streptococcal pharyngitis, pulmonary tuberculo- BDProbeTec strand displacement amplification (SDA) system for
sis, vaginitis, chlamydial and gonococcal infections. MLO Med Lab diagnosis of tuberculosis in non-respiratory and respiratory samples.
Obs 2004;36:10 – 5. J Med Microbiol 2004;53:1215 – 9.
[4] Smith K, Diggle MA, Clarke SC. Automation of a fluorescence- [22] Cosentino LA, Landers DV, Hillier SL. Detection of Chlamydia
based multiplex PCR for the laboratory confirmation of common trachomatis and Neisseria gonorrhoeae by strand displacement
bacterial pathogens. J Med Microbiol 2004;53:115 – 7. amplification and relevance of the amplification control for use with
[5] Nakaguchi Y, Ishizuka T, Ohnaka S, et al. Rapid and specific vaginal swab specimens. J Clin Microbiol 2003;41:3592 – 6.
detection of tdh, trh1, and trh2 mRNA of Vibrio parahaemolyticus [23] Demidov VV. Rolling-circle amplification in DNA diagnostics: the
by transcription-reverse transcription concerted reaction with an power of simplicity. Expert Rev Mol Diagn 2002;2:542 – 8.
automated system. J Clin Microbiol 2004;42:4284 – 92. [24] Louie L, Matsumura SO, Choi E, et al. Evaluation of three rapid
[6] Bankowski MJ, Anderson SM. Real-time nucleic acid amplification methods for detection of methicillin resistance in Staphylococcus
in clinical microbiology. Clin Microbiol Newsl 2004;26:9 – 15. aureus. J Clin Microbiol 2000;38:2170 – 3.
[7] Apfalter P, Assadian O, Blasi F, et al. Reliability of nested PCR for [25] Zheng X, Kolbert CP, Varga-Delmore P, et al. Direct mecA detection
detection of Chlamydia pneumoniae DNA in atheromas: results from from blood culture bottles by branched-DNA signal amplification.
a multicenter study applying standardized protocols. J Clin Microbiol J Clin Microbiol 1999;37:4192 – 3.
2002;40:4428 – 34. [26] Van Der Pol B, Williams JA, Smith NJ, et al. Evaluation of the
[8] Yam WC, Cheng VC, Hui WT, et al. Direct detection of Digene Hybrid Capture II Assay with the Rapid Capture System for
Mycobacterium tuberculosis in clinical specimens using single-tube detection of Chlamydia trachomatis and Neisseria gonorrhoeae.
biotinylated nested polymerase chain reaction-enzyme linked J Clin Microbiol 2002;40:3558 – 64.
immunoassay (PCR-ELISA). Diagn Microbiol Infect Dis 2004;48: [27] Girdner JL, Cullen AP, Salama TG, et al. Evaluation of the
271 – 275. digene hybrid capture II CT-ID test for detection of Chlamydia
[9] Boyadzhyan B, Yashina T, Yatabe JH, et al. Comparison of the trachomatis in endocervical specimens. J Clin Microbiol 1999;
APTIMA CT and GC assays with the APTIMA combo 2 assay, the 37:1579 – 81.
Abbott LCx assay, and direct fluorescent-antibody and culture assays [28] Wellinghausen N, Wirths B, Essig A, Wassill L. Evaluation of the
for detection of Chlamydia trachomatis and Neisseria gonorrhoeae. Hyplex BloodScreen Multiplex PCR-Enzyme-linked immunosorbent
J Clin Microbiol 2004;42:3089 – 93. assay system for direct identification of gram-positive cocci and
[10] Keer JT, Birch L. Molecular methods for the assessment of bacterial gram-negative bacilli from positive blood cultures. J Clin Microbiol
viability. J Microbiol Methods 2003;53:175 – 83. 2004;42:3147 – 52.
[11] Loens K, Ieven M, Ursi D, et al. Detection of Mycoplasma [29] Lee CY, Panicker G, Bej AK. Detection of pathogenic bacteria in
pneumoniae by real-time nucleic acid sequence-based amplification. shellfish using multiplex PCR followed by CovaLink NH microwell
J Clin Microbiol 2003;41:4448 – 50. plate sandwich hybridization. J Microbiol Methods 2003;53:199 – 209.
[12] Mahony JB, Song X, Chong S, et al. Evaluation of the NucliSens [30] Taha MK. Simultaneous approach for nonculture PCR-based
Basic Kit for detection of Chlamydia trachomatis and Neisseria identification and serogroup prediction of Neisseria meningitidis.
gonorrhoeae in genital tract specimens using nucleic acid J Clin Microbiol 2000;38:855 – 7.
sequence-based amplification of 16S rRNA. J Clin Microbiol [31] Spiro A, Lowe M, Brown D. A bead-based method for multiplexed
2001;39:1429 – 35. identification and quantitation of DNA sequences using flow
[13] Rodriguez-Lazaro D, Lloyd J, Herrewegh A, et al. A molecular cytometry. Appl Environ Microbiol 2000;66:4258 – 65.
beacon-based real-time NASBA assay for detection of Mycobacte- [32] Dunbar SA, Vander Zee CA, Oliver KG, et al. Quantitative,
rium avium subsp. paratuberculosis in water and milk. FEMS multiplexed detection of bacterial pathogens: DNA and protein
Microbiol Lett 2004;237:119 – 26. applications of the Luminex LabMAP system. J Microbiol Methods
[14] Rodriguez-Lazaro D, D’Agostino M, Pla M, Cook N. Construction 2003;53:245 – 52.
strategy for an internal amplification control for real-time diag- [33] Exner MM, Lewinski MA. Sensitivity of multiplex real-time PCR
nostic assays using nucleic acid sequence-based amplification: reactions, using the LightCycler and the ABI PRISM 7700 Sequence
development and clinical application. J Clin Microbiol 2004;42: Detection System, is dependent on the concentration of the DNA
5832 – 6. polymerase. Mol Cell Probes 2002;16:351 – 7.
[15] Hill CS. Molecular diagnostic testing for infectious diseases using [34] Giglio S, Monis PT, Saint CP. Demonstration of preferential binding
TMA technology. Expert Rev Mol Diagn 2001;1:445 – 55. of SYBR Green I to specific DNA fragments in real-time multiplex
[16] Gaydos CA, Theodore M, Dalesio N, et al. Comparison of three PCR. Nucleic Acids Res 2003;31:e136.
nucleic acid amplification tests for detection of Chlamydia tracho- [35] Hamels S, Gala JL, Dufour S. et al. Consensus PCR and microarray
matis in urine specimens. J Clin Microbiol 2004;42:3041 – 5. for diagnosis of the genus Staphylococcus, species, and methicillin
[17] Lemaitre N, Armand S, Vachee A, et al. Comparison of the real-time resistance. Biotechniques 2001; 31:1364 – 1366, 1368, 1370 – 1372.
PCR method and the Gen-Probe amplified Mycobacterium tuberculosis [36] Barlaan EA, Sugimori M, Furukawa S, Takeuchi K. Electronic
direct test for detection of Mycobacterium tuberculosis in pulmonary microarray analysis of 16S rDNA amplicons for bacterial detection.
and nonpulmonary specimens. J Clin Microbiol 2004;42:4307 – 9. J Biotechnol 2005;115:11 – 21.
[18] Gamboa F, Fernandez G, Padilla E, et al. Comparative evaluation of [37] Roth SB, Jalava J, Ruuskanen O, et al. Use of an oligonucleotide
initial and new versions of the Gen-Probe amplified Mycobacterium array for laboratory diagnosis of bacteria responsible for acute upper
tuberculosis direct test for direct detection of Mycobacterium respiratory infections. J Clin Microbiol 2004;42:4268 – 74.
218 E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220

[38] Yu X, Susa M, Knabbe C, et al. Development and validation of a [58] Diggle MA, Clarke SC. Genotypic characterization of Neisseria
diagnostic DNA microarray to detect quinolone-resistant Escherichia meningitidis using pyrosequencing trade mark. Mol Biotechnol
coli among clinical isolates. J Clin Microbiol 2004;42:4083 – 91. 2004;28:139 – 46.
[39] Patel JB. 16S rRNA gene sequencing for bacterial pathogen [59] Unemo M, Olcen P, Jonasson J, Fredlund H. Molecular typing of
identification in the clinical laboratory. Mol Diagn 2001;6:313 – 21. Neisseria gonorrhoeae isolates by pyrosequencing of highly poly-
[40] Kiratisin P, Li L, Murray PR, Fischer SH. Identification of bacteria morphic segments of the porB gene. J Clin Microbiol 2004;42:
recovered from clinical specimens by 16S rRNA gene sequencing. 2926 – 34.
Eur J Clin Microbiol Infect Dis 2003;22:628 – 31. [60] Jonasson J, Olofsson M, Monstein HJ. Classification, identification
[41] Chen CC, Teng LJ, Chang TC. Identification of clinically relevant and subtyping of bacteria based on pyrosequencing and signature
viridans group streptococci by sequence analysis of the 16S – 23S matching of 16S rDNA fragments. Apmis 2002;110:263 – 72.
ribosomal DNA spacer region. J Clin Microbiol 2004;42:2651 – 7. [61] Ronaghi M, Elahi E. Pyrosequencing for microbial typing.
[42] Fischer M, Whitney A, Ahmed S, Barry J, Jones J. Unexplained J Chromatogr B Analyt Technol Biomed Life Sci 2002;782:67 – 72.
severe illness and death among injecting drug users in Scotland, [62] Poppert S, Essig A, Marre R, et al. Detection and differentiation of
Ireland, and England from April to August 2000. Washington (DC)’ chlamydiae by fluorescence in situ hybridization. Appl Environ
ASM Press; 2004. Microbiol 2002;68:4081 – 9.
[43] Nilsson WB, Paranjype RN, DePaola A, Strom MS. Sequence [63] Jensen TK, Boye M, Ahrens P, et al. Diagnostic examination of
polymorphism of the 16S rRNA gene of Vibrio vulnificus is a human intestinal spirochetosis by fluorescent in situ hybridization
possible indicator of strain virulence. J Clin Microbiol 2003;41: for Brachyspira aalborgi, Brachyspira pilosicoli, and other
442 – 6. species of the genus Brachyspira (Serpulina). J Clin Microbiol
[44] Sacchi CT, Whitney AM, Reeves MW, et al. Sequence diversity of 2001;39:4111 – 8.
Neisseria meningitidis 16S rRNA genes and use of 16S rRNA gene [64] Jansen GJ, Mooibroek M, Idema J, et al. Rapid identification of
sequencing as a molecular subtyping tool. J Clin Microbiol 2002;40: bacteria in blood cultures by using fluorescently labeled oligonucleo-
4520 – 7. tide probes. J Clin Microbiol 2000;38:814 – 7.
[45] Sacchi CT, Whitney AM, Mayer LW, et al. Sequencing of 16S rRNA [65] Trebesius K, Adler K, Vieth M, et al. Specific detection and
gene: a rapid tool for identification of Bacillus anthracis. Emerg prevalence of Helicobacter heilmannii like organisms in the human
Infect Dis 2002;8:1117 – 23. gastric mucosa by fluorescent in situ hybridization and partial 16S
[46] Cook VJ, Turenne CY, Wolfe J, et al. Conventional methods versus ribosomal DNA sequencing. J Clin Microbiol 2001;39:1510 – 6.
16S ribosomal DNA sequencing for identification of nontuberculous [66] Trebesius K, Leitritz L, Adler K, et al. Culture independent
mycobacteria: cost analysis. J Clin Microbiol 2003;41:1010 – 5. and rapid identification of bacterial pathogens in necrotising
[47] Hall L, Doerr KA, Wohlfiel SL, Roberts GD. Evaluation of the fasciitis and streptococcal toxic shock syndrome by fluorescence
MicroSeq system for identification of mycobacteria by 16S riboso- in situ hybridisation. Med Microbiol Immunol (Berl) 2000;188:
mal DNA sequencing and its integration into a routine clinical 169 – 75.
mycobacteriology laboratory. J Clin Microbiol 2003;41:1447 – 53. [67] Kempf VA, Trebesius K, Autenrieth IB. Fluorescent in situ hybrid-
[48] Woo PC, Ng KH, Lau SK, et al. Usefulness of the MicroSeq 500 16S ization allows rapid identification of microorganisms in blood
ribosomal DNA-based bacterial identification system for identifica- cultures. J Clin Microbiol 2000;38:830 – 8.
tion of clinically significant bacterial isolates with ambiguous [68] Nielsen PE, Egholm M, Berg RH, Buchardt O. Sequence-selective
biochemical profiles. J Clin Microbiol 2003;41:1996 – 2001. recognition of DNA by strand displacement with a thymine-
[49] Gee JE, De BK, Levett PN. Use of 16S rRNA gene sequencing for substituted polyamide. Science 1991;254:1497 – 500.
rapid confirmatory identification of Brucella isolates. J Clin Micro- [69] Egholm M, Buchardt O, Christensen L, et al. PNA hybridizes to
biol 2004;42:3649 – 54. complementary oligonucleotides obeying the Watson – Crick hydro-
[50] Gee JE, Sacchi CT, Glass MB, et al. Use of 16S rRNA gene gen-bonding rules. Nature 1993;365:566 – 8.
sequencing for rapid identification and differentiation of Burkholde- [70] Stender H. PNA FISH: an intelligent stain for rapid diagnosis of
ria pseudomallei and B. mallei. J Clin Microbiol 2003;41:4647 – 54. infectious diseases. Expert Rev Mol Diagn 2003;3:649 – 55.
[51] Clarridge III JE. Impact of 16S rRNA gene sequence analysis for [71] Oliveira K, Procop GW, Wilson D, et al. Rapid identification of
identification of bacteria on clinical microbiology and infectious Staphylococcus aureus directly from blood cultures by fluorescence
diseases. Clin Microbiol Rev 2004;17:840 – 62. in situ hybridization with peptide nucleic acid probes. J Clin
[52] Harmsen D, Rothganger J, Frosch M, Albert J. RIDOM: ribosomal Microbiol 2002;40:247 – 51.
differentiation of medical micro-organisms database. Nucleic Acids [72] Oliveira K, Haase G, Kurtzman C, et al. Differentiation of Candida
Res 2002;30:416 – 7. albicans and Candida dubliniensis by fluorescent in situ hybrid-
[53] Schuurman T, de Boer RF, Kooistra-Smid AM, van Zwet AA. ization with peptide nucleic acid probes. J Clin Microbiol 2001;39:
Prospective study of use of PCR amplification and sequencing of 4138 – 41.
16S ribosomal DNA from cerebrospinal fluid for diagnosis of [73] Rigby S, Procop GW, Haase G, et al. Fluorescence in situ hybrid-
bacterial meningitis in a clinical setting. J Clin Microbiol 2004;42: ization with peptide nucleic acid probes for rapid identification of
734 – 40. Candida albicans directly from blood culture bottles. J Clin Micro-
[54] Wong DA, Yip PC, Tse DL, et al. Routine use of a simple low- biol 2002;40:2182 – 6.
cost genotypic assay for the identification of mycobacteria in a [74] Gonzalez V, Padilla E, Gimenez M, et al. Rapid diagnosis of
high throughput laboratory. Diagn Microbiol Infect Dis 2003;47: Staphylococcus aureus bacteremia using S. aureus PNA FISH. Eur J
421 – 6. Clin Microbiol Infect Dis 2004;23:396 – 8.
[55] Kim WS, Khunajakr N, Ren J, Dunn NW. Conservation of the major [75] Chapin K, Musgnug M. Evaluation of three rapid methods for the
cold shock protein in lactic acid bacteria. Curr Microbiol 1998;37: direct identification of Staphylococcus aureus from positive blood
333 – 6. cultures. J Clin Microbiol 2003;41:4324 – 7.
[56] Ronaghi M, Uhlen M, Nyren P. A sequencing method based on real- [76] Stender H, Oliveira K, Rigby S, et al. Rapid detection, identi-
time pyrophosphate. Science 1998;281:363 – 5. fication, and enumeration of Escherichia coli by fluorescence in
[57] Monstein H, Nikpour-Badr S, Jonasson J. Rapid molecular identi- situ hybridization using an array scanner. J Microbiol Methods
fication and subtyping of Helicobacter pylori by pyrosequencing of 2001;45:31 – 9.
the 16S rDNA variable V1 and V3 regions. FEMS Microbiol Lett [77] Esiobu N, Mohammed R, Echeverry A, et al. The application of
2001;199:103 – 7. peptide nucleic acid probes for rapid detection and enumeration of
E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220 219

eubacteria, Staphylococcus aureus and Pseudomonas aeruginosa in [97] Burggraf S, Olgemoller B. Simple technique for internal control of
recreational beaches of S. Florida. J Microbiol Methods 2004;57: real-time amplification assays. Clin Chem 2004;50:819 – 25.
157 – 62. [98] Vet JA, Marras SA. Design and optimization of molecular beacon
[78] Brandt O, Feldner J, Stephan A, et al. PNA microarrays for real-time polymerase chain reaction assays. Methods Mol Biol
hybridisation of unlabelled DNA samples. Nucleic Acids Res 2005;288:273 – 90.
2003;31:e119. [99] Nikkari S, McLaughlin IJ, Bi W, et al. Does blood of healthy
[79] Kambhampati D, Nielsen PE, Knoll W. Investigating the kinetics of subjects contain bacterial ribosomal DNA? J Clin Microbiol
DNA – DNA and PNA – DNA interactions using surface plasmon 2001;39:156 – 9.
resonance-enhanced fluorescence spectroscopy. Biosens Bioelectron [100] Black CM, Marrazzo J, Johnson RE, et al. Head-to-head multi-
2001;16:1109 – 18. center comparison of DNA probe and nucleic acid amplification
[80] Wang J. PNA biosensors for nucleic acid detection. Curr Issues Mol tests for Chlamydia trachomatis infection in women performed
Biol 1999;1:117 – 22. with an improved reference standard. J Clin Microbiol 2002;40:
[81] Takiya T, Futo S, Tsuna M, et al. Identification of single base-pair 3757 – 63.
mutation on uidA gene of Escherichia coli O157:H7 by Peptide [101] Martin DH, Cammarata C, Van Der Pol B, et al. Multicenter
Nucleic Acids (PNA) mediated PCR clamping. Biosci Biotechnol evaluation of AMPLICOR and automated COBAS AMPLICOR
Biochem 2004;68:360 – 8. CT/NG tests for Neisseria gonorrhoeae. J Clin Microbiol 2000;38:
[82] Xi C, Balberg M, Boppart SA, Raskin L. Use of DNA and peptide 3544 – 9.
nucleic acid molecular beacons for detection and quantification of [102] Akduman D, Ehret JM, Messina K, et al. Evaluation of a strand
rRNA in solution and in whole cells. Appl Environ Microbiol displacement amplification assay (BD ProbeTec-SDA) for detection
2003;69:5673 – 8. of Neisseria gonorrhoeae in urine specimens. J Clin Microbiol
[83] Wolffs P, Knutsson R, Sjoback R, Radstrom P. PNA-based light-up 2002;40:281 – 3.
probes for real-time detection of sequence0specific PCR products. [103] Sloutsky A, Han LL, Werner BG. Practical strategies for
Biotechniques 2001; 31:766, 769 – 771. performance optimization of the enhanced Gen-Probe amplified
[84] Cirillo DM, Piana F, Frisicale L, et al. Direct rapid diagnosis of Mycobacterium tuberculosis direct test. J Clin Microbiol 2004;42:
rifampicin-resistant M. tuberculosis infection in clinical samples by 1547 – 51.
line probe assay (INNO LiPA Rif-TB). New Microbiol 2004;27: [104] Piersimoni C, Scarparo C, Piccoli P, et al. Performance assessment of
221 – 7. two commercial amplification assays for direct detection of Myco-
[85] Jureen P, Werngren J, Hoffner SE. Evaluation of the line probe assay bacterium tuberculosis complex from respiratory and extrapulmo-
(LiPA) for rapid detection of rifampicin resistance in Mycobacterium nary specimens. J Clin Microbiol 2002;40:4138 – 42.
tuberculosis. Tuberculosis (Edinb) 2004;84:311 – 6. [105] Warren DK, Liao RS, Merz LR, et al. Detection of methicillin-
[86] Miller N, Infante S, Cleary T. Evaluation of the LiPA resistant Staphylococcus aureus directly from nasal swab speci-
MYCOBACTERIA assay for identification of mycobacterial mens by a real-time PCR assay. J Clin Microbiol 2004;42:
species from BACTEC 12B bottles. J Clin Microbiol 2000;38: 5578 – 81.
1915 – 1919. [106] Chapin KC, Blake P, Wilson CD. Performance characteristics and
[87] Mijs W, De Vreese K, Devos A, et al. Evaluation of a commercial utilization of rapid antigen test, DNA probe, and culture for detection
line probe assay for identification of Mycobacterium species from of group a streptococci in an acute care clinic. J Clin Microbiol
liquid and solid culture. Eur J Clin Microbiol Infect Dis 2002;21: 2002;40:4207 – 10.
794 – 802. [107] Davies HD, Miller MA, Faro S, et al. Multicenter study of a
[88] Lindholm L, Sarkkinen H. Direct identification of gram-positive rapid molecular-based assay for the diagnosis of group B
cocci from routine blood cultures by using AccuProbe tests. J Clin Streptococcus colonization in pregnant women. Clin Infect Dis
Microbiol 2004;42:5609 – 13. 2004;39:1129 – 35.
[89] Gut IG. DNA analysis by MALDI-TOF mass spectrometry. Hum [108] Briselden AM, Hillier SL. Evaluation of affirm VP Microbial
Mutat 2004;23:437 – 41. Identification Test for Gardnerella vaginalis and Trichomonas
[90] Lowe CA, Diggle MA, Clarke SC. A single nucleotide poly- vaginalis. J Clin Microbiol 1994;32:148 – 52.
morphism identification assay for the genotypic characterisation of [109] Brown HL, Fuller DD, Jasper LT, et al. Clinical evaluation of affirm
Neisseria meningitidis using MALDI-TOF mass spectrometry. Br J VPIII in the detection and identification of Trichomonas vaginalis,
Biomed Sci 2004;61:8 – 10. Gardnerella vaginalis, and Candida species in vaginitis/vaginosis.
[91] von Wintzingerode F, Bocker S, Schlotelburg C, et al. Base-specific Infect Dis Obstet Gynecol 2004;12:17 – 21.
fragmentation of amplified 16S rRNA genes analyzed by mass [110] Bukhari Z, Weihe J, LeChevallier M. Development of procedures for
spectrometry: a tool for rapid bacterial identification. Proc Natl Acad rapid detection of E. coli O157:H7 from source and finished water
Sci U S A 2002;99:7039 – 44. samples. Water Sci Technol 2004;50:233 – 7.
[92] Lefmann M, Honisch C, Bocker S, et al. Novel mass spectrometry- [111] Levett PN, Morey RE, Galloway RL, et al. Detection of pathogenic
based tool for genotypic identification of mycobacteria. J Clin leptospires by real-time quantitative PCR. J Med Microbiol
Microbiol 2004;42:339 – 46. 2005;54:45 – 9.
[93] Radstrom P, Knutsson R, Wolffs P, et al. Pre-PCR processing: [112] Mothershed EA, Sacchi CT, Whitney AM, et al. Use of real-time
strategies to generate PCR-compatible samples. Mol Biotechnol PCR to resolve slide agglutination discrepancies in serogroup
2004;26:133 – 46. identification of Neisseria meningitidis. J Clin Microbiol
[94] Millar BC, Xu J, Moore JE. Risk assessment models and contam- 2004;42:320 – 8.
ination management: implications for broad-range ribosomal DNA [113] Tomaso H, Scholz HC, Al Dahouk S, et al. Development of 5V
PCR as a diagnostic tool in medical bacteriology. J Clin Microbiol nuclease real-time PCR assays for the rapid identification of the
2002;40:1575 – 80. burkholderia mallei//burkholderia pseudomallei complex. Diagn Mol
[95] Borst A, Box AT, Fluit AC. False-positive results and contami- Pathol 2004;13:247 – 53.
nation in nucleic acid amplification assays: suggestions for a [114] Kosters K, Reischl U, Schmetz J, et al. Real-time LightCycler PCR
prevent and destroy strategy. Eur J Clin Microbiol Infect Dis for detection and discrimination of Bordetella pertussis and
2004;23:289 – 99. Bordetella parapertussis. J Clin Microbiol 2002;40:1719 – 22.
[96] Aslanzadeh J. Preventing PCR amplification carryover contamina- [115] Reischl U, Youssef MT, Wolf H, et al. Real-time fluorescence PCR
tion in a clinical laboratory. Ann Clin Lab Sci 2004;34:389 – 96. assays for detection and characterization of heat-labile I and heat-
220 E.A. Mothershed, A.M. Whitney / Clinica Chimica Acta 363 (2006) 206 – 220

stable I enterotoxin genes from enterotoxigenic Escherichia coli. [131] Ribault S, Harper K, Grave L, et al. Rapid screening method for
J Clin Microbiol 2004;42:4092 – 100. detection of bacteria in platelet concentrates. J Clin Microbiol
[116] Belanger SD, Boissinot M, Clairoux N, et al. Rapid detection of 2004;42:1903 – 8.
Clostridium difficile in feces by real-time PCR. J Clin Microbiol [132] Jacobs MR, Bajaksouzian S, Windau A, et al. Evaluation of the
2003;41:730 – 4. Scansystem method for detection of bacterially contaminated
[117] Ferris MJ, Masztal A, Martin DH. Use of species-directed 16S rRNA platelets. Transfusion 2005;45:265 – 9.
gene PCR primers for detection of Atopobium vaginae in patients [133] Wang R, Tombelli S, Minunni M, et al. Immobilisation of DNA
with bacterial vaginosis. J Clin Microbiol 2004;42:5892 – 4. probes for the development of SPR-based sensing. Biosens Bioelec-
[118] Hoffmaster AR, Ravel J, Rasko DA, et al. Identification of anthrax tron 2004;20:967 – 74.
toxin genes in a Bacillus cereus associated with an illness [134] Malicka J, Gryczynski I, Gryczynski Z, Lakowicz JR. Use of surface
resembling inhalation anthrax. Proc Natl Acad Sci U S A 2004; plasmon-coupled emission to measure DNA hybridization. J Biomol
101:8449 – 54. Screen 2004;9:208 – 15.
[119] Woo PC, Lau SK, Woo GK, et al. Bacteremia due to Clostridium [135] Wang R, Minunni M, Tombelli S, Mascini M. A new approach for
hathewayi in a patient with acute appendicitis. J Clin Microbiol the detection of DNA sequences in amplified nucleic acids by a
2004;42:5947 – 9. surface plasmon resonance biosensor. Biosens Bioelectron
[120] Westergren V, Bassiri M, Engstrand L. Bacteria detected by culture 2004;20:598 – 605.
and 16S rRNA sequencing in maxillary sinus samples from intensive [136] Hofstadler SA, Sampath R, Blyn LB, et al. TIGER: the universal
care unit patients. Laryngoscope 2003;113:270 – 5. biosensor. Int J Mass Spectrom 2005;242:23 – 41.
[121] Lau SK, Woo PC, Mok MY, et al. Characterization of Haemophilus [137] Graham DL, Ferreira HA, Freitas PP. Magnetoresistive-based
segnis, an important cause of bacteremia, by 16S rRNA gene biosensors and biochips. Trends Biotechnol 2004;22:455 – 62.
sequencing. J Clin Microbiol 2004;42:877 – 80. [138] Ji HF, Yang X, Zhang J, Thundat T. Molecular recognition of
[122] Kignel S, Almeida Pina F, Andre E, et al. Occurrence of Helicobacter biowarfare agents using micromechanical sensors. Expert Rev Mol
pylori in dental plaque and saliva of dyspeptic patients. Oral Dis Diagn 2004;4:859 – 66.
2005;11:17 – 21. [139] Mannelli I, Minunni M, Tombelli S, Mascini M. Quartz crystal
[123] Pottumarthy S, Schapiro JM, Prentice JL, et al. Clinical isolates of microbalance (QCM) affinity biosensor for genetically modified
Staphylococcus intermedius masquerading as methicillin-resistant organisms (GMOs) detection. Biosens Bioelectron 2003;18:129 – 40.
Staphylococcus aureus. J Clin Microbiol 2004;42:5881 – 4. [140] Deisingh AK, Thompson M. Sequences of E. coli O157:H7 detected
[124] Saglani S, Harris KA, Wallis C, Hartley JC. Empyema: the use of by a PCR-acoustic wave sensor combination. Analyst 2001;126:
broad range 16S rDNA PCR for pathogen detection. Arch Dis Child 2153 – 8.
2005;90:70 – 3. [141] Kim DS, Jeong YT, Park HJ, et al. An FET-type charge sensor for
[125] Higgins JA, Nasarabadi S, Karns JS, et al. A handheld real time highly sensitive detection of DNA sequence. Biosens Bioelectron
thermal cycler for bacterial pathogen detection. Biosens Bioelectron 2004;20:69 – 74.
2003;18:1115 – 23. [142] Uslu F, Ingebrandt S, Mayer D, et al. Label-free fully electronic
[126] Emanuel PA, Bell R, Dang JL, et al. Detection of Francisella nucleic acid detection system based on a field-effect transistor device.
tularensis within infected mouse tissues by using a hand-held PCR Biosens Bioelectron 2004;19:1723 – 31.
thermocycler. J Clin Microbiol 2003;41:689 – 93. [143] Gau JJ, Lan EH, Dunn B, et al. A MEMS based amperometric
[127] Healy M, Huong J, Bittner T, et al. Microbial DNA typing by detector for E. coli bacteria using self-assembled monolayers.
automated repetitive-sequence-based PCR. J Clin Microbiol Biosens Bioelectron 2001;16:745 – 55.
2005;43:199 – 207. [144] Kai E, Ikebukuro K, Hoshina S, et al. Detection of PCR products of
[128] Lising M, Reece K, Walton D, et al. Identification of gram-negative Escherichia coli O157:H7 in human stool samples using surface
bacteria using the Diversilab system. 104th General Meeting. New plasmon resonance (SPR). FEMS Immunol Med Microbiol 2000;29:
Orleans (LA)’ American Society for Microbiology; 2004. 283 – 8.
[129] Webb R, Lising M, Reece K, Healy M. Identification and strain [145] Gulliksen A, Solli L, Karlsen F, et al. Real-time nucleic acid
differentiation of Bacillus species using automated rep-PCR DNA sequence-based amplification in nanoliter volumes. Anal Chem
fingerprinting. ASM Biodefense Research Meeting. Baltimore, MD’ 2004;76:9 – 14.
American Society for Microbiology; 2003.
[130] Pathak S, Choi SK, Arnheim N, Thompson ME. Hydroxylated
quantum dots as luminescent probes for in situ hybridization. J Am
Chem Soc 2001;123:4103 – 4.

Anda mungkin juga menyukai