Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

SUPPLEMENT ARTICLE

Molecular Diagnostics for the Detection


and Characterization of Microbial Pathogens
Gary W. Procop

Downloaded from https://academic.oup.com/cid/article/45/Supplement_2/S99/285931 by guest on 03 August 2021


Department of Pathology, Jackson Memorial Hospital and University of Miami Miller School of Medicine, Miami, Florida

New and advanced methods of molecular diagnostics are changing the way we practice clinical microbiology,
which affects the practice of medicine. Signal amplification and real-time nucleic acid amplification technologies
offer a sensitive and specific result with a more rapid turnaround time than has ever before been possible.
Numerous methods of postamplification analysis afford the simultaneous detection and differentiation of
numerous microbial pathogens, their mechanisms of resistance, and the construction of disease-specific assays.
The technical feasibility of these assays has already been demonstrated. How these new, often more expensive
tests will be incorporated into routine practice and the impact they will have on patient care remain to be
determined. One of the most attractive uses for such techniques is to achieve a more rapid characterization
of the infectious agent so that a narrower-spectrum antimicrobial agent may be used, which should have an
impact on resistance patterns.

It has been 12 decades since the inception of the PCR lecular revolution” under way. Although this is exciting,
[1, 2]. Although this technique was almost immediately the responsibility of the laboratorian and the ordering
implemented in research laboratories to study a variety physician is to ensure that these tests are used appro-
of infectious diseases, among other processes, it has priately. The abuse of this technology has serious im-
been within only the past 5–7 years that these assays plications for cost as well as patient care, because, as
have been made so user-friendly that they may be im- with any test, false-positive and false-negative reactions
plemented in any microbiology laboratory [3]. In ad- may occur.
dition to PCR, there are numerous competitive tech- The clinical microbiologist is occasionally asked to
nologies that have been developed and shown to be exclude the presence of a particular microorganism in
useful for the detection and characterization of micro- a clinical specimen. For example, CSF may be submitted
organisms. Many of these more recently developed as- for the exclusion of herpes simplex virus, or a blood
says may be performed by medical technologists who, smear may be submitted for the exclusion of Plasmo-
although highly skilled, do not have advanced training dium species. However, in many instances, several tests
in molecular biology. The simplification of this type of are ordered per clinical specimen, and the questions
testing means that it may be performed after hours or asked are of a broader nature. For example, when a
in adverse conditions (i.e., in the field). Several of these blood or wound culture is ordered, the question really
assays are US Food and Drug Administration approved is “Are there any bacteria or fungi present in this clinical
or cleared, and more are in this process. Today, in the specimen, and to what antimicrobial agents are they
modern microbiology laboratory, there truly is a “mo- susceptible?” The clinician rarely draws a blood sample
for culture for the detection or exclusion of a single
type of microorganism (i.e., blood is not drawn for
Reprints or correspondence: Dr. Gary W. Procop, Dept. of Pathology, University culture to “rule out [r/o] Pseudomonas” but, rather, to
of Miami Miller School of Medicine, Jackson Memorial Hospital (R-5), 1611 NW
12th Ave., Holtz Bldg., Rm. 2090, Miami, FL 33136 (gprocop@med.miami.edu). “r/o bacteremia and fungemia”). The task of the clinical
Clinical Infectious Diseases 2007; 45:S99–111 microbiologist is complicated, because he or she must
 2007 by the Infectious Diseases Society of America. All rights reserved.
1058-4838/2007/4505S2-0002$15.00
be aware of all of the potential pathogens that may be
DOI: 10.1086/519259 present in a clinical specimen and must devise or utilize

Molecular Diagnostics • CID 2007:45 (Suppl 2) • S99


means by which to detect and characterize these organisms. culture samples [17–21]. Another technology, target-capture
The molecular diagnostic techniques used for the detection technology, used predominantly for the detection of viruses
and characterization of microorganisms may be separated into (HPV, cytomegalovirus, and hepatitis B virus), could also be
several broad categories. These are direct hybridization, nucleic used for the detection of bacteria and fungi in positive blood
acid amplification, and a variety of methods for postamplifi- culture samples [22–25].
cation analysis. These methods may be used to either detect or In situ hybridization. In situ hybridization has been in-
exclude particular pathogens or, more usefully, to assess a spec- troduced into the clinical microbiology laboratory and should
imen or positive culture sample for a wide variety of micro- prove to be a useful technology for the rapid characterization
organisms. The value of such applications must be rigorously of bacteria and fungi in positive blood culture samples, which
assessed, because these tests are usually added to the battery of is an area of particular interest to many [5, 7, 9–11, 13, 15,
routinely performed assays. The benefits of such assays are less 26–30]. In situ hybridization has also been used successfully in

Downloaded from https://academic.oup.com/cid/article/45/Supplement_2/S99/285931 by guest on 03 August 2021


likely to be found in the laboratory and more likely to be seen direct clinical specimens and in histologic sections for the de-
by the clinician or the health care administrator. For example, tection of microorganisms [8, 12, 14]. Regardless of the type
the more rapid detection and characterization of an infecting of direct hybridization technology used, identifications using
pathogen afford the clinician the opportunity to tailor anti- signal hybridizations are most accurately obtained after micro-
microbial therapy. In addition to benefiting the patients, this organism growth or biological amplification of the organism.
may save health care dollars through the pharmacy by iden- This technology is often used to differentiate microorganisms
tifying situations wherein less expensive antimicrobial agents of similar morphotypes in positive culture samples (e.g., gram-
may be used [4, 5]. For example, Forrest et al. [5] demonstrated positive cocci in clusters or acid-fast bacilli). One of the ad-
that $1729 in pharmacy costs per patient could be saved by vantages of this technology is that it may be used in conjunction
switching the patient’s treatment from a more expensive echi- with traditional methods, such as Gram staining. The infor-
nocandin to a less expensive azole. The use of narrower-spec- mation derived from Gram staining may be used to select the
trum agents, in contrast to the use of broad-spectrum agents, appropriate probes from a battery to more specifically address
should also slow the selection for and spread of antimicrobial- select questions. However, the use of the full battery of probes
resistant microorganisms. Other benefits that have been noted on all blood culture bottles that indicate positivity without the
include better use of ancillary diagnostic tests (e.g., radiology Gram stain–derived information would be cost-prohibitive and
services) and of hospital beds (i.e., decreasing lengths of stay an inefficient use of resources. The information derived from
or the number of unnecessary admissions) [6]. Gram staining is used to select the one or few probes necessary
for bacterial or fungal characterization. For example, if gram-
DIRECT HYBRIDIZATION positive cocci in clusters are demonstrated in the Gram stain,
There are many direct-hybridization techniques used in clinical then one would select a probe that would detect Staphylococcus
microbiology. Some of these have been used for the rapid iden- aureus and differentiate it from coagulase-negative staphylo-
tification and characterization of bacteria and fungi in blood cocci. Similarly, a battery of probes for the most commonly
culture samples, whereas others have been used for different occurring yeast species would be used when yeasts are seen in
applications (e.g., human papillomavirus [HPV] detection) but the Gram stain, whereas probes for Pseudomonas and members
could also potentially be used in this manner. In situ hybrid- of the Enterobacteriaceae would be used when gram-negative
ization will predominate this discussion, because it is the newest bacilli are seen in the Gram stain smear. This is an excellent
signal amplification technique to be used in the clinical mi- example of optimizing the use of new technology through the
crobiology laboratory. The detection process used in in situ use of established methods. A variety of oligonucleotide probe
hybridization may be fluorescent (FISH) or chromogenic sequences have already been described that generate family-,
(CISH). These tools, which were once used solely in the do- genus-, and species-specific information [13, 15].
mains of researchers and anatomic pathologists, have been Two types of oligonucleotide probes are most frequently used
demonstrated to be useful for the detection of pathogens in for in situ hybridization. These are traditional nucleic acid ol-
blood culture samples as well as in other clinical specimens [7– igonucleotide probes and peptide nucleic acid (PNA) probes.
16]. A chemiluminescence-generating chemical reaction used Traditional oligonucleotides used for in situ hybridization need
after the direct hybridization of oligonucleotide probes to mi- no further explanation for most in the field of molecular di-
croorganism ribosomal RNA (rRNA) is another technology that agnostics, but a limited description of PNA probes is warranted
may be used for the rapid characterization of microorganisms. [31, 32]. Peptide nucleic acid probes are synthetic molecules
This technology has been used for more than a decade for the that do not naturally occur. The most important difference
identification of microorganisms, such as commonly occurring between DNA and PNA resides in the “backbone” of the mol-
Mycobacterium species and the dimorphic fungi, in positive ecule. The backbone of the DNA polymer consists of repeating

S100 • CID 2007:45 (Suppl 2) • Procop


deoxyribose sugar and phosphate molecules, the latter of which to identify the most common causes of infection in patients
have a net negative charge. In contrast, the backbone of the with cystic fibrosis, to differentiate mycobacteria in culture and
PNA molecule, used in the studies described here, consists of in histologic sections, and to detect trypanosomes in patients
repeating glycine molecules. Glycine, importantly, is a sterically with African sleeping sickness [8, 14, 27].
small and neutrally charged amino acid. The similarity between The same hybridization probes used in the clinical micro-
DNA and PNA oligonucleotide probes is the 4 common nu- biology laboratory could be used in in situ hybridization re-
cleotide bases, which participate in Watson and Crick base actions in histologic sections. Hayden et al. have demonstrated,
pairing. These unique properties afford the construction of a in a series of articles [12, 33–35], that chromogenic in situ
probe molecule that can penetrate through the cell wall and hybridization may be used successfully to differentiate mor-
cell membrane (i.e., a hydrophobic lipid bilayer) of microor- phologically similar microorganisms. They showed the feasi-
ganisms while retaining the specificity achieved with Watson bility of this technology to detect Legionella pneumophila and

Downloaded from https://academic.oup.com/cid/article/45/Supplement_2/S99/285931 by guest on 03 August 2021


and Crick base pairing [32]. The charge of the in situ probe is differentiate filamentous bacteria (i.e., aerobic actinomyces),
particularly important in microbiology, wherein the microor- filamentous fungi, and yeast and yeastlike fungi. Importantly,
ganism undergoing FISH analysis is intact, and in distinct con- they showed the ability to separate the systemic dimorphic
trast to many applications in molecular pathology (e.g., FISH fungi, which appear as yeast (e.g., Histoplasma capsulatum and
for Her-2 amplification), wherein the nucleus, which contains Blastomyces dermatitidis) or spherules (e.g., Coccidioides im-
the chromosomal targets for the FISH assay, has been sectioned, mitis), from Candida and Cryptococcus species. Although it may
and the probes are applied directly onto their nucleic acid tar- seem a morphologically simple task to differentiate a well-de-
gets (i.e., penetration through a lipid bilayer is not necessary). veloped spherule of C. immitis from budding yeast, it is con-
Although there is limited commercial availability of FISH
siderably more complicated when intact spherules are not seen
probes in microbiology, some are available, and more are likely
and, rather, only immature spherules and endospores are pres-
on the horizon. The PNA FISH assays that are currently avail-
ent. In situ hybridization has also been used to identify Pneu-
able in North America detect and differentiate S. aureus from
mocystis jiroveci [36]. Finally, one can imagine an economic
other gram-positive cocci in clusters and Candida albicans from
benefit when the same probe sets are used for both microbi-
other yeasts. The assay may be performed as soon as the blood
ology and infectious disease pathology applications. For ex-
culture bottle indicates positivity and Gram staining has been
ample, PNA FISH probes have also been used successfully to
performed, or the slides to be tested may be tested in batches.
identify mycobacteria in histologic sections and from positive
Although batch testing clearly increases the time to result that
culture samples [27].
is possible with immediate testing, it is a viable alternative for
Another exciting application that may have practical uses in
laboratories with limited personnel and still has a better time
the clinical microbiology laboratory is the combined use of in
to result than traditional methods. It is now possible, with this
situ hybridization and flow cytometry. Small-scale flow cyto-
technology, to provide a definitive species-level identification
meters that can detect microorganisms have been developed
of microorganisms, such as S. aureus and C. albicans, on the
and are routinely used in the food and pharmaceutical indus-
same day that the blood culture bottle indicates positivity and
tries. These applications often use nonspecific dyes that bind
demonstrates gram-positive cocci in clusters or yeast, respec-
tively [7, 9–11, 26, 30]. In addition to the commercially avail- to the external aspect of the bacteria. Although such assays are
able applications, several studies have demonstrated the feasi- useful for determining overall bacterial counts in a product,
bility of definitively identifying other medically important the results generated do not yield genus- or species-level in-
bacteria and yeast species [13, 15, 28, 29]. Importantly, these formation. However, it has been demonstrated that this tech-
assays are simple to perform and represent a molecular diag- nology can be used to detect microorganisms to which in situ
nostic tool that may be introduced into laboratories without probes are hybridized [37, 38]. This combination of technol-
previous experience in molecular microbiology. They have al- ogies affords the replacement of human interpretation, which
ready been adopted and are performed routinely in the several is subjective and requires advanced training and experience to
clinical microbiology laboratories in the United States. From perfect, with objective, quantitative measures that are per-
an economic standpoint, the use of the C. albicans PNA FISH formed by an instrument.
probe has been shown to be cost-effective, particularly when The precise role that in situ hybridization will play in the
linked to limiting the use of more expensive antifungal drugs, clinical microbiology laboratory and its impact on patient care
such as an echinocandin, when a less expensive but equally remain to be seen. Several other competitive technologies, some
efficacious drug like fluconazole is a viable alternative [4, 5]. of which are described below, offer similar advantages with
In addition to its use in the differentiation of the causes of regard to rapid identification. Regardless, it has been demon-
bacteremia and fungemia, in situ hybridization has been used strated that the identification of the most common microor-

Molecular Diagnostics • CID 2007:45 (Suppl 2) • S101


ganisms that cause bacteremia and fungemia may be rapidly in conjunction with quantitative standards, these could be used
determined by use of this technology. to provide quantitative information (e.g., viral loads). The com-
Other methods of signal amplification. There are several bination of ⭓2 nucleic acid amplification reactions into 1 assay
other signal-amplification methods that are currently used in is a multiplex reaction. These assays have been used to detect
many clinical microbiology laboratories, the applications of multiple pathogens simultaneously but require advanced design
which could be expanded. The chemiluminescent rRNA probe and critical assessments, because intramolecular interactions
hybridization technology that has been available from Gen- between the various primers and probes are potential limita-
Probe for years could be expanded/promoted to detect many tions of such assays. Several multiplex (and monoplex) assays
of these pathogens. This technology uses probes that target are commercially available as “analyte-specific reagents.” These
rRNA, which is present in numerous copies in each cell. The are commercial products that contain all the necessary com-
hybridized probe is then detected using a proprietary chemical ponents to conduct a particular amplification reaction, which

Downloaded from https://academic.oup.com/cid/article/45/Supplement_2/S99/285931 by guest on 03 August 2021


reaction that results in the generation of light. Products that are manufactured under Good Manufacturing Practice con-
utilize this technology have been used for years in many lab- ditions, but the responsibility of validating such assays resides
oratories for the rapid identification of the most commonly with the testing laboratories. One of the more successful of the
occurring mycobacteria and for isolates suspected to represent multiplex analyte-specific reagents that has been released is a
systemic dimorphic fungi [18–21, 26, 39–41]. Although prod- real-time RT-PCR that simultaneously detects and differenti-
ucts have been developed for rapid detection of many of the ations the most important respiratory viral pathogens—influ-
more commonly occurring bacteria, such as S. aureus, these enza A, influenza B, and respiratory syncytial virus. The real-
have not gained widespread adoption for the rapid identifi- time version currently available is a truncated version of
cation of the common causes of bacteremia and fungemia [26, another multiplex assay produced by the same company that
42, 43]. There have, however, been some hints that this com- detected and differentiated 6 respiratory viruses [49, 50].
pany might be interested in developing panels useful for the Broad-range nucleic acid amplification is another approach
identification of the commonly encountered bacteria and yeast that could be used to detect any of a wide variety of micro-
species, although a commercial product has not become organisms are in a taxonomically related group [51–54]. The
available. results of broad-range PCR simply indicate that one of the
The hybrid capture (Digene) technology, which has been members of a particular group is present. Quantitative data
used most successfully for the detection of high-risk HPV sub- (i.e., organism load) may also be obtained if quantitative stan-
types, is also a signal amplification technology [44]. This tech- dards are included in the amplification run and a standard
nology has also been used for the detection of cytomegalovirus, curve has been produced. Although quantitative data may be
hepatitis B virus, Neisseria gonorrhoeae, and Chlamydia tra-
of use, much of the strength of broad-range nucleic acid am-
chomatis [45–47]. Although signal amplification assays that do
plification assays comes from the information that may be de-
not employ direct microscopy are often not as sensitive as
rived from the amplified product (i.e., the amplicon) through
nucleic acid amplification, some have shown the contrary [44,
postamplification analysis. There are many methods of post-
48]. These assays have adequate sensitivity for many clinical
amplification analysis, but only a few will be briefly discussed
applications and afford high specificity, depending on the spec-
here, given the scope of the present article. These are postam-
ificity of the probes used. In addition to the types of applications
plification melt curve analysis, reverse hybridization, DNA se-
described, this technology could also be used to characterize
quencing (traditional and pyrosequencing), and solid- and liq-
bacteria or fungi that grow in culture and require further
uid-phase microarray analysis.
identification.
It seems that advances in laboratory diagnostics more com-
monly occur quite rapidly in conjunction with technological
NUCLEIC ACID AMPLIFICATION
advances (i.e., as in Stephen J. Gould’s theory of punctuated
Nucleic acid amplification includes not only PCR but also al- equilibrium), rather than occurring though the accumulation
ternate technologies, such as strand-displacement amplification of small advances over a long period (i.e., a more Darwinian-
and transcription-mediated amplification (i.e., nucleic acid se- type change) [55, 56]. The history of the use of PCR in clinical
quence–based amplification), which have also proven useful in laboratories supports this notion. Although PCR and other
clinically important assays. In addition to the various chemical nucleic acid amplification technologies (note: hereafter, “PCR”
reactions that may be used for amplification, the design of these will be used generically to encompass all methods of nucleic
assays varies widely. Monoplex assays are amplification reac- acid amplification) have penetrated into the larger diagnostic
tions that detect a single target and provide a “present or ab- and university laboratories for the detection of microorganisms
sent”–type result. For example, a Legionella pneumophila–spe- that are usually difficult to culture, they have not until recently
cific PCR could be used to rapidly detect this pathogen. If used become readily available in small, perhaps less molecularly

S102 • CID 2007:45 (Suppl 2) • Procop


savvy laboratories or for the detection of more common path- the signal that is detected during the PCR is evidence that the
ogens. This is because traditional PCR is cumbersome and slow, target DNA molecule is present, because the oligonucleotide
especially in light of the time and process necessary to specif- probe hybridizes to its target through specific Watson and Crick
ically determine the nature of the amplified product by South- base-pairing rules. There are many different types of oligo-
ern blot analysis. However, all this changed with the advent of nucleotide probes available. The 3 most commonly used types
real-time PCR, also known as “homogeneous, rapid cycle nu- are the hydrolysis or “TaqMan” probes, molecular beacons, and
cleic acid amplification.” fluorescence resonance energy transfer (FRET) probes. A pos-
The advent of real-time PCR awaited complementary ad- itive real-time PCR result for any of these probes is demon-
vances in engineering and chemistry. Engineers devised ways strated by an exponential increase in fluorescence with respect
of decreasing the PCR cycling time. This has been accomplished to amplification time or cycle number (figure 1). Specimens
through the use of heated and cooled air to achieve more rapid that do not contain the target molecule (i.e., that are negative)

Downloaded from https://academic.oup.com/cid/article/45/Supplement_2/S99/285931 by guest on 03 August 2021


heating and cooling, respectively. This is in contrast to tradi- will demonstrate minimal or no change in fluorescence. The
tional block cycle heating, wherein, traditionally, a metal or qualitative information derived from such reactions is useful
solid block was heated and cooled to change the temperatures for demonstrating the presence or absence of a microorganism
of the reaction vessels contained in the block to achieve those of interest. For example, assays have been devised to address
temperatures necessary for the PCR. This latter method, al- the presence or absence of Legionella pneumophila by use of
though effective, is naturally slower and reflects the physical real-time PCR. Such an assay may offer times to results that
differences between conduction and convection heating. Next, are similar to those of direct immunofluorescence antigen de-
the engineers needed to develop sufficient optical systems for tection, but the real-time PCR has been shown to be more
these instruments, to both introduce a beam of light of a par- sensitive than direct immunofluorescence antigen detection;
ticular wavelength and detect minute changes in light emitted additionally, the same assay was described as being more rapid
from the solution within the reaction vessel. Fortunately, many and possibly more sensitive than culture while retaining a spec-
of these technical issues had been addressed by scientists who ificity equivalent to that of culture [58].
used laser and fluorescent dye technology for various appli- The specificity of a PCR assay is determined by the target
cations, such as flow cytometry. This afforded the adaptation DNA sequence under evaluation, the sequence of the oligo-
of such technology to real-time PCR platforms. nucleotide probe, similar sequences that may exist elsewhere
The advances in chemistry that made real-time PCR possible in nature, and the intentions of the assay designer. Many assays
were also significant, and modifications of these chemical re- are species specific, but genus-generic assays (i.e., a more broad-
range PCR) may also be designed. These would be useful when
actions continue today. The challenge was to devise a way that
it is important to detect all members of a group that may be
amplification and detection reactions could occur within the
divergent in nature. For example, one may use a species-specific
same reaction vessel without necessitating the opening of the
PCR for Salmonella serotype Typhi, to detect that particular
vessel and the manipulation of the amplified product. Main-
serotype, or a broad-range Salmonella assay, to detect all of the
taining a closed system is important in a clinical microbiology
members of that genus [59].
laboratory, because chemical contamination of the environ-
In addition to the detection of microbial pathogens, these
ment with amplicon could lead to false-positive reactions [57].
applications may be used to detect genetic elements that encode
This was achieved through the discovery and construction of
for mechanisms of antimicrobial resistance (e.g., the mecA
molecules that were nonfluorescent in a nonhybridized state
gene), those that determine subtypes of such genes for epi-
but were able to generate a fluorescent signal when both bound
demiologic purposes (e.g., the mecA cassette IV associated with
to their DNA target and excited by the appropriate electro-
community-acquired Staphylococcus aureus infection), and the
magnetic energy. The signal generated may be nonspecific,
detection of virulence factors (e.g., Panton-Valentine leukoci-
demonstrating only the presence of double-stranded DNA (i.e.,
din). Although useful for the detection of certain mechanisms
evidence that the PCR has occurred), or specific, which is
of resistance, nucleic acid amplification assays for more com-
achieved through the hybridization of specific, fluorescently
plicated and genetically diverse mechanisms of antimicrobial
labeled oligonucleotide probes. An example of a nonspecific
resistance (e.g., the extended-spectrum b-lactamases) have re-
probe is SYBR Green dye I, which binds to the minor groove
mained elusive and are not likely to replace traditional phe-
of double-stranded DNA. The signal derived from this mole-
notypic susceptibility testing in the near future.
cule, while PCR is under way (thus, in real time), demonstrates
to the user that amplification is occurring. Whether this am-
POSTAMPLIFICATION ANALYSIS
plification is specific cannot be determined at this point in the
reaction (see the melt curve analysis below). Conversely, if one Melt curve analysis. Another feature of some of the chemical
uses a specific, fluorescently labeled oligonucleotide probe, then reactions used with real-time PCR is the ability to perform a

Molecular Diagnostics • CID 2007:45 (Suppl 2) • S103


Downloaded from https://academic.oup.com/cid/article/45/Supplement_2/S99/285931 by guest on 03 August 2021
Figure 1. Examples of real-time PCR amplification reactions. The negative control specimen has produced a flat line, whereas the positive control
and other template-containing samples demonstrate an exponential increase in fluorescence. The reactions that become positive earlier (i.e., that have
a lower crossing point, or cycle number threshold [Ct]) contain a greater amount of target template than do those that became positive later.

postamplification melt curve analysis. This provides different be detected in the instrument’s “quantification” mode, and, if
information, depending on the type of probe used. If SYBR such a scenario occurred, the specimen would erroneously be
Green dye I or another nonspecific DNA binding dye is used, deemed positive. However, if a postamplification melt curve
then the information derived is similar to that derived from analysis was performed, then the nucleotide mismatches be-
gel electrophoresis. Larger amplicons are held together by more tween the probe and target sequences would be detected, be-
hydrogen bonds, whereas smaller amplicons, such as primer cause the probe could not hybridize to its target sequence with
dimers, are held together by fewer hydrogen bonds. The post- the optimal affinity and would “melt off” at a lower temper-
amplification melt curve derived using these types of DNA ature. This technology is so sensitive that it is commonly used
binding molecules demonstrates high melting temperatures to determine SNPs in human genetic pathology (e.g., the SNP
(95C–98C) for large amplicons and lower temperatures for responsible for factor V Leiden) [61]. Thus, postamplification
smaller amplicons and primer dimers. Some researchers have melt curve analysis is a method whereby one may obtain ad-
demonstrated the detection and differentiation of single nu- ditional data concerning the specific nature of the amplified
cleotide polymorphisms (SNPs) by use of high-resolution melt DNA product after real-time PCR.
curves and nonspecific DNA binding dyes, but, for the most This difference in melt curves created by imperfect comple-
part, these uses remain experimental [60]. mentarity between probes and their target hybridization sites
The FRET probes and another oligonucleotide probe called may be exploited to differentiate taxonomically related micro-
an “eclipse probe” are the types of specific oligonucleotide organisms to the species level. Assays have been designed to
probes that provide the best postamplification melt curves. differentiate HSV type 1 and 2, the BK and JC polyoma viruses,
Foremost, a melt curve analysis after amplification provides human herpesvirus 6 types A and B, the commonly occurring
information regarding the specificity of the reaction. A partic- Bartonella species, and M. tuberculosis from nontuberculous
ular melting temperature is expected for the hybridization be- mycobacteria (figure 2), among others [53, 62]. This applica-
tween the oligonucleotide probe and its target. The presence tion is quite useful, but there is some evidence that there may
of the expected melt curve after amplification helps to assure be diminished sensitivity for the target that has the lower melt-
the user that the results are correct. If, for example, the primers ing temperature (i.e., the imperfect complementarity) [63].
were degenerate and also amplified a related microorganism, Therefore, it would be prudent to not use this type of appli-
the melt curve generated would have a lower temperature than cation if the target that had this profile caused serious disease
expected, if nucleotide polymorphisms existed at the probe (i.e., if there would be a significant sequela associated with a
hybridization site. This molecular misidentification would not false-negative reaction) or if such a target organism is present

S104 • CID 2007:45 (Suppl 2) • Procop


Downloaded from https://academic.oup.com/cid/article/45/Supplement_2/S99/285931 by guest on 03 August 2021
Figure 2. Postamplification melt curves generated after a broad-range PCR for mycobacteria. DNA extracts from Mycobacterium tuberculosis (TB)
and nontuberculous mycobacteria (NTM) are used as controls. The melt curve derived from a specimen that contained mycobacteria is clearly categorized
here as NTM, which means that the patient does not require respiratory isolation or antituberculous therapy.

in small quantities and may be missed because of the limited hybridization, DNA sequencing—including pyrosequencing—
sensitivity. The approach we have taken with the mycobacteria and microarray analysis, with a focus on limited microarrays
has been to utilize this technology to differentiate mycobacteria of both the solid and liquid formats.
that are detected either in the acid-fast smear or in histologic There is often a need to assess for or identify 11 microbial
sections stained by the Ziehl-Neelsen method. In such an in- pathogen. It is more time- and cost-effective if these assess-
stance, it is not a question of “if” mycobacteria are present but, ments can be performed simultaneously or in a single reaction
rather, a question of “which type of” mycobacteria is present, than if they are performed in numerous monoplex amplifi-
with the primary goal being the separation of M. tuberculosis cation reactions. For example, it is necessary to assay for nu-
from the nontuberculous mycobacteria (figure 2). The hybrid- merous high-risk HPVs to appropriately diagnose or exclude
ization probes have 100% homology to M. tuberculosis, with a high-risk HPV infection in a woman. It is preferable to per-
the nontuberculous mycobacteria having a ⭓1-nt mismatch at form a single assay to detect and/or differentiate these viruses
the probe hybridization site. This design makes the assay most rather than to perform 110 individual PCRs for the various
sensitive for the detection of the most important mycobacteria high-risk HPV subtypes. Postamplification analyses are often
that we encounter in North American microbiology, M. tu- employed when the number of results obtained exceeds the
berculosis. If acid-fast bacilli are seen in the smear or stained technical differentiation capabilities of multiplex reactions
tissue section, but amplified product is not detected, then one wherein each reaction contains a different fluorescent label, or
may categorize the result as “quantity of mycobacteria below when the number of results achieved may exceed the differ-
the limit of detection of this assay,” which is a more useful entiation capabilities of a melt curve analysis.
result than a false-negative result. Reverse hybridization. One of the most simple-to-perform
There are a variety of other methods of postamplification methods of detecting a variety of pathogens, after either a mul-
amplicon analysis that require the opening and manipulation tiplex or a broad-range amplification reaction, is through re-
of amplified PCR product. These are acceptable methods, but verse hybridization. This technique is the opposite of Southern
care must be taken to avoid the introduction of amplified prod- blot analysis in many aspects and is therefore termed “reverse.”
uct into the areas of master mix preparation or specimen prep- Herein, a variety of probes are placed on a nitrocellulose
aration, to avoid false-positive reactions. The types of methods membrane strip, and the amplicon rather than the probe is
are too numerous to describe in an exhaustive manner in this labeled. The amplicon is applied to the strip under appropriate
article, so I have selected a few to highlight that either have or hybridization conditions, and then wash steps are performed.
will likely have an impact in clinical medicine; these are reverse If the amplicon hybridizes to one of the immobilized probes,

Molecular Diagnostics • CID 2007:45 (Suppl 2) • S105


it may be visualized through the application of a color devel-
opment reagent. The position of the product on the strip reveals
the nature of the pathogen. This technology has been used to
detect and differentiate the commonly occurring mycobacteria
(figure 3), clinically important fungi, HPV subtypes, hepatitis
C virus genotypes, and certain resistance-associated mutations
in HIV [64–67]. This technology is simple to use, but some
applications have been limited because of the expense of the
product. Occasionally, light bands of nonspecific hybridization
are seen (i.e., “ghost bands”) that may confuse interpretation
or suggest dual infections.

Downloaded from https://academic.oup.com/cid/article/45/Supplement_2/S99/285931 by guest on 03 August 2021


DNA sequencing: traditional (Sanger) and pyrosequencing.
The sequencing of the amplicon, through traditional Sanger
sequencing, is the ultimate in postamplification analysis. The
entire composition of the amplified product is thereby deter-
mined. Although initially difficult to perform, this technology
is now readily available to any molecular diagnostic laboratory.
Advances in capillary and gel electrophoresis, as well as com-
puter-assisted sequence analysis, afford the expanded use of
this technology to many more laboratories. Traditionally, se-
quencing after RT-PCR is the method of choice for determining
resistance-associated mutations in HIV, and 2 Food and Drug
Administration–approved tests have been developed for this
application. This technology is also commonly used to identify
bacteria, fungi, and mycobacteria on the basis of sequences
within the ribosomal gene complex [68–71]. Once the sequence
of an unknown microorganism is determined, it may be queried Figure 3. Reverse-hybridization test demonstrating a single postam-
plification assay that could be used to differentiate the majority of my-
against a genetic database that contains thousands of sequences.
cobacteria that are clinically encountered. Here, all of the hybridization
The results of a query are returned as a percentage match, which sites are developed. The only lines that would be present if a clinical
assists the molecular microbiologist with the identification of specimen or a positive culture sample were tested that contained a single
the microorganisms. There is clearly great promise for this species of mycobacteria would be the lines that correspond to the species,
approach, and commercial products (e.g., MicroSeq [Applied the Mycobacterium genus, and the conjugate control position. The ab-
breviations on the left side of the reverse hybridization strip correlate
Biosystems]) and nonpublic databases (e.g., SmartGene) have
with the species or groups on the right side of the strip. MAIS,
been established. Full-length sequencing is particularly pow- Mycobacterium avium–M. intracellulare–M. scrofulaceum; MTB, M.
erful when several nucleotide polymorphisms are needed for tuberculosis.
the characterization of a microbe and these are distant from
one another within the amplicon (e.g., some of the mutations incorporated, then the light generated will be twice that gen-
associated with HIV resistance). The drawbacks associated with erated by the incorporation of a single nucleotide (e.g., …G).
this technology would include the cost of the equipment, re- The instrument records the dispensation of the nucleotides and
agents, and access to private databases; the need to analyze long the light generated. Nucleotides that are dispensed but not
lengths of sequenced DNA, which is time and labor consuming; incorporated are hydrolyzed by apyrase, one of the enzymes in
and the inability to generate high-quality sequence just down- the mixture. Hereby, the DNA sequence is determined as it is
stream of the sequencing primer hybridization site. being synthesized. The strengths of this technology are that it
Another type of DNA sequencing that has been more recently is user friendly, the reactions occur in the commonly used 96-
described is sequencing by synthesis, or pyrosequencing [72, well plate, and the sequences immediately downstream of the
73]. This technology uses proprietary chemical reactions in- sequencing primer are readily determined, which is ideal for
cluding several enzymes that generate light when a nucleotide SNP analysis. The limitations of this approach include the cost
is incorporated into the strand of DNA that is being synthe- of the instrumentation and reagents, its limited ability to ap-
sized. The amount of this light is in proportion to the pyro- propriately determine the sequence of homopolymers (i.e., nu-
phosphate released during nucleotide incorporation. For ex- merous nucleotides of the same type occurring one after an-
ample, if 2 of the same nucleotides (e.g., …GG) are other), and the length of high-quality DNA sequence that may

S106 • CID 2007:45 (Suppl 2) • Procop


be generated, which is !100 nt and frequently is !50 nt. As examined, so that normal genetic variation may be understood
noted, this technology is particularly useful for SNP analysis, and incorporated into identification schema (i.e., some varia-
because the assessment of only a limited number of nucleotides tion may reflect only strain variation, whereas other variation
is necessary. In addition, this technology, although perhaps not is taxonomically meaningful for the differentiation of species).
as discriminating as Sanger sequencing, may be used in the The bottom line is that the sequence-based identification of
taxonomic categorization of clinically relevant microorganisms. microorganisms is only as good as the sequences in the genetic
For this application, it is necessary to determine the sequence library used and the characterization of the strains from which
of variable regions that contain unique or “signature” sequences these sequences were derived.
for microorganisms within a group. This technology has been Microarray technology. Microarray technology has proven
used to appropriately categorize mycobacteria and nocardiae an important tool for discovery [80–82]. These platforms may
into clinically important groups (e.g., M. tuberculosis complex) be used to assay for numerous (i.e., hundreds) of targets si-

Downloaded from https://academic.oup.com/cid/article/45/Supplement_2/S99/285931 by guest on 03 August 2021


[74]. In our experience at the Cleveland Clinic, this technique multaneously and have been a critical means of studying the
of identifying mycobacteria was less expensive than our tra- expression of multiple genes simultaneously. This technology,
ditional approach that utilized genetic probes and biochemical regardless of its technical format, consists of numerous indi-
reactions (data not shown). It has been used to identify yeasts vidual probe-target hybridization reactions that are assayed for
(figure 4), filamentous fungi, and a variety of bacteria [75, 76]. simultaneously. Not surprisingly, this technology has been used
It has also been used to differentiate the BK and JC polyoma to differentiate numerous microbial pathogens, study their ex-
viruses, as well as HPV subtypes [77–79]. pression profiles, and detect the genetic determinants of drug
Regardless of the type of sequencing technology used, both resistance [83–86]. For example, soon after its introduction, it
the laboratorian performing the sequencing and the ordering was shown to be capable of differentiating medically important
clinician must be aware of the strengths and limitations of these mycobacteria and detecting the most common causes of resis-
assays. In addition, the identifications achieved using these tance to antimycobacterial drugs [87, 88]. Unfortunately, the
methods are only as sound as the entries in the genetic databases limitations of this technology, such as cost, inconsistent reac-
that are being queried. Genetic database entries should be based tions in some systems, and data management and interpreta-
on isolates that have been soundly identified using traditional tion, have surpassed its practical uses. From a practical stand-
methods (if feasible) and should be of good sequence quality. point, simply more data are generated from a single, large array
Unfortunately, this is not always the case. Finally, an adequate than are necessary in clinical medicine, and the cost of the array
number of well-identified isolates from each species should be is prohibitive for routine use. Fortunately, this is changing. One

Figure 4. Pyrogram (i.e., tracing derived from pyrosequencing) of a 40-bp region that has been used for the identification of the most commonly
occurring yeast species. The sizes of the peaks are measurements of the amount of light generated, which correlates to the amount of pyrophosphate
generated during nucleotide incorporation in the process of DNA synthesis. C. parapsilosis, Candida parapsilosis.

Molecular Diagnostics • CID 2007:45 (Suppl 2) • S107


Downloaded from https://academic.oup.com/cid/article/45/Supplement_2/S99/285931 by guest on 03 August 2021
Figure 5. Limited or small-scale bioelectric microarray demonstrating the feasibility of this type of technology to differentiate most of the clinically
important mycobacteria. A Mycobacterium genus site is located on the far side of the microarray, whereas the remainder is occupied by species-
specific or complex-specific (e.g., Mycobacterium tuberculosis complex) hybridization sites. ITS, internal transcribed spacer region.

of the most promising areas in molecular diagnostics is the use identify the typical causes of invasive fungal and mycobacterial
of limited microarrays for the assessment of numerous genetic infections (figure 5). This approach is optimized with the in-
targets after either a multiplex reaction or a broad-range PCR. clusion of broad family or kingdom probes. For example, a
A number of different microarray platforms have been de- Raoutella species is an uncommon cause of bacteremia, and
scribed, including bioelectric arrays and liquid microarrays. As genus- or species-level probe sites are not likely to be included
with any technology, there are strengths and limitations with in a “bacteremia” microarray. The presence of an uncommon
each system. These systems used today have improved hybrid- bacterium such as this in the blood culture sample could be
ization and reporting for the individual probe/target combi- detected through the inclusion of a broad-range bacterial probe
nations (i.e., they have been made more reliable), and, perhaps site on the microarray. Hybridization at this site, with the ab-
most importantly, some have been scaled down from large sence of hybridization at a species-determining site, would
platforms to smaller-sized versions that address clinically im- demonstrate the presence of a bacterium other than any of the
portant issues that require multiple results. In clinical micro- more common causes of bacteremia that are included in the
biology, a microarray with 20–30 hybridization sites would be panel.
sufficient to address the most common causes of bacteremia The possibility of producing disease-specific microassays also
and fungemia (i.e., the most common causes of positive blood exists, because these formats may be used after broad-range
culture results), and similarly sized arrays could be used to PCR, multiplex PCR, or a combination of such assays. For

S108 • CID 2007:45 (Suppl 2) • Procop


example, one could envision a microarray that could distin- 6. Ramers C, Billman G, Hartin M, Ho S, Sawyer MH. Impact of a
diagnostic cerebrospinal fluid enterovirus polymerase chain reaction
guish between the various causes of meningoencephalitis. Such test on patient management. JAMA 2000; 283:2680–5.
an assay may include a complex multiplex reaction that con- 7. Oliveira K, Brecher SM, Durbin A, et al. Direct identification of Staph-
sisted of a broad-range bacterial PCR, a species-specific PCR ylococcus aureus from positive blood culture bottles. J Clin Microbiol
2003; 41:889–91.
for Cryptococcus neoformans, a broad-range RT-PCR for the
8. Radwanska M, Magez S, Perry-O’Keefe H, et al. Direct detection and
enteroviruses, and a specific PCR for HSV. Although not ex- identification of African trypanosomes by fluorescence in situ hybrid-
haustive, such an array would, in a single postamplification ization with peptide nucleic acid probes. J Clin Microbiol 2002; 40:
assay, address all the most common etiologic agents causing 4295–7.
9. Rigby S, Procop GW, Haase G, et al. Fluorescence in situ hybridization
meningoencephalitis. Furthermore, additional probe sites could with peptide nucleic acid probes for rapid identification of Candida
be added, as needed. For example, a West Nile virus site could albicans directly from blood culture bottles. J Clin Microbiol 2002; 40:
be added, if desired. Limited microarrays, from a business per- 2182–6.

Downloaded from https://academic.oup.com/cid/article/45/Supplement_2/S99/285931 by guest on 03 August 2021


10. Oliveira K, Procop GW, Wilson D, Coull J, Stender H. Rapid identi-
spective, represent a potentially disruptive technology, because fication of Staphylococcus aureus directly from blood cultures by fluo-
the widespread use of these could change the way that we rescence in situ hybridization with peptide nucleic acid probes. J Clin
practice microbiology. However, their use will depend on nu- Microbiol 2002; 40:247–51.
11. Oliveira K, Haase G, Kurtzman C, Hyldig-Nielsen JJ, Stender H. Dif-
merous factors, such as the development of useful products,
ferentiation of Candida albicans and Candida dubliniensis by fluores-
cost, Food and Drug Administration approval, reimbursement, cent in situ hybridization with peptide nucleic acid probes. J Clin
and, most importantly, their performance in the clinical lab- Microbiol 2001; 39:4138–41.
oratory and their impact on patient care. 12. Hayden RT, Uhl JR, Qian X, et al. Direct detection of Legionella species
from bronchoalveolar lavage and open lung biopsy specimens: com-
In summary, molecular methods are changing the way that parison of LightCycler PCR, in situ hybridization, direct fluorescence
we practice laboratory medicine and thereby are affecting the antigen detection, and culture. J Clin Microbiol 2001; 39:2618–26.
practice of clinical medicine. New methods of direct hybridi- 13. Kempf VA, Trebesius K, Autenrieth IB. Fluorescent in situ hybridization
allows rapid identification of microorganisms in blood cultures. J Clin
zation, rapid-cycle homogeneous PCR, and a variety of pos- Microbiol 2000; 38:830–8.
tamplification methods of analysis are making the genetic-based 14. Hogardt M, Trebesius K, Geiger AM, Hornef M, Rosenecker J, Heese-
identification and characterization of pathogenic microorgan- mann J. Specific and rapid detection by fluorescent in situ hybridization
of bacteria in clinical samples obtained from cystic fibrosis patients. J
isms more rapid and, in many instances, more accurate than
Clin Microbiol 2000; 38:818–25.
ever before. However, for each of these technologies, the ad- 15. Jansen GJ, Mooibroek M, Idema J, Harmsen HJ, Welling GW, Degener
ditions to health care costs must be weighed against the po- JE. Rapid identification of bacteria in blood cultures by using fluo-
tential advantages of more rapid diagnostics. The techniques rescently labeled oligonucleotide probes. J Clin Microbiol 2000; 38:
814–7.
and the technologies are here. Well-controlled outcome stud- 16. Drobniewski FA, More PG, Harris GS. Differentiation of Mycobacte-
ies are now needed to demonstrate the efficacy of these rium tuberculosis complex and nontuberculous mycobacterial liquid
technologies. cultures by using peptide nucleic acid-fluorescence in situ hybridization
probes. J Clin Microbiol 2000; 38:444–7.
17. Scarparo C, Piccoli P, Rigon A, Ruggiero G, Nista D, Piersimoni C.
Acknowledgments
Direct identification of mycobacteria from MB/BacT alert 3D bottles:
Supplement sponsorship. This article was published as part of a sup- comparative evaluation of two commercial probe assays. J Clin Mi-
plement entitled “Annual Conference on Antimicrobial Resistance,” spon- crobiol 2001; 39:3222–7.
sored by the National Foundation for Infectious Diseases. 18. Louro AP, Waites KB, Georgescu E, Benjamin WH Jr. Direct identi-
Potential conflicts of interest. G.W.P. is a scientific advisor for Lu- fication of Mycobacterium avium complex and Mycobacterium gordonae
minex, AdvanDX, Roche, Cepheid, bioMerieux, and BD GeneOhm and from MB/BacT bottles using AccuProbe. J Clin Microbiol 2001; 39:
receives royalties for licensing from Biotage and BD GeneOhm. 570–3.
19. Alcaide F, Benitez MA, Escriba JM, Martin R. Evaluation of the BAC-
TEC MGIT 960 and the MB/BacT systems for recovery of mycobacteria
References
from clinical specimens and for species identification by DNA
1. Mullis K, Faloona F, Scharf S, Saiki R, Horn G, Erlich H. Specific AccuProbe. J Clin Microbiol 2000; 38:398–401.
enzymatic amplification of DNA in vitro: the polymerase chain reac- 20. Beggs ML, Stevanova R, Eisenach KD. Species identification of My-
tion. 1986. Biotechnology 1992; 24:17–27. cobacterium avium complex isolates by a variety of molecular tech-
2. Shampo MA, Kyle RA. Kary B. Mullis—Nobel Laureate for procedure niques. J Clin Microbiol 2000; 38:508–12.
to replicate DNA. Mayo Clin Proc 2002; 77:606. 21. Richter E, Niemann S, Rusch-Gerdes S, Hoffner S. Identification of
3. Espy MJ, Uhl JR, Sloan LM, et al. Real-time PCR in clinical micro- Mycobacterium kansasii by using a DNA probe (AccuProbe) and mo-
biology: applications for routine laboratory testing. Clin Microbiol Rev lecular techniques. J Clin Microbiol 1999; 37:964–70.
2006; 19:165–256. 22. Hesselink AT, Bulkmans NW, Berkhof J, Lorincz AT, Meijer CJ, Snijders
4. Alexander BD, Ashley ED, Reller LB, Reed SD. Cost savings with im- PJ. Cross-sectional comparison of an automated hybrid capture 2 assay
plementation of PNA FISH testing for identification of Candida al- and the consensus GP5+/6+ PCR method in a population-based cer-
bicans in blood cultures. Diagn Microbiol Infect Dis 2006; 54:277–82. vical screening program. J Clin Microbiol 2006; 44:3680–5.
5. Forrest GN, Mankes K, Jabra-Rizk MA, et al. Peptide nucleic acid 23. Hui CK, Bowden S, Zhang HY, et al. Comparison of real-time PCR
fluorescence in situ hybridization-based identification of Candida al- assays for monitoring serum hepatitis B virus DNA levels during an-
bicans and its impact on mortality and antifungal therapy costs. J Clin tiviral therapy. J Clin Microbiol 2006; 44:2983–7.
Microbiol 2006; 44:3381–3. 24. Chemaly RF, Yen-Lieberman B, Castilla EA, et al. Correlation between

Molecular Diagnostics • CID 2007:45 (Suppl 2) • S109


viral loads of cytomegalovirus in blood and bronchoalveolar lavage in primary cervical screening: a systematic review and meta-analysis
specimens from lung transplant recipients determined by histology and of non-randomized studies. Gynecol Oncol 2007; 104:232–46.
immunohistochemistry. J Clin Microbiol 2004; 42:2168–72. 45. Ho SK, Li FK, Lai KN, Chan TM. Comparison of the CMV Brite Turbo
25. Chan HL, Leung NW, Lau TC, Wong ML, Sung JJ. Comparison of assay and the Digene Hybrid Capture CMV DNA (version 2.0) assay
three different sensitive assays for hepatitis B virus DNA in monitoring for quantitation of cytomegalovirus in renal transplant recipients. J
of responses to antiviral therapy. J Clin Microbiol 2000; 38:3205–8. Clin Microbiol 2000; 38:3743–5.
26. Chapin K, Musgnug M. Evaluation of three rapid methods for the 46. Konnick EQ, Erali M, Ashwood ER, Hillyard DR. Evaluation of the
direct identification of Staphylococcus aureus from positive blood cul- COBAS amplicor HBV monitor assay and comparison with the ultra-
tures. J Clin Microbiol 2003; 41:4324–7. sensitive HBV hybrid capture 2 assay for quantification of hepatitis B
27. Lefmann M, Schweickert B, Buchholz P, et al. Evaluation of peptide virus DNA. J Clin Microbiol 2005; 43:596–603.
nucleic acid-fluorescence in situ hybridization for identification of clin- 47. Van Der Pol B, Williams JA, Smith NJ, et al. Evaluation of the Digene
ically relevant mycobacteria in clinical specimens and tissue sections. Hybrid Capture II Assay with the Rapid Capture System for detection
J Clin Microbiol 2006; 44:3760–7. of Chlamydia trachomatis and Neisseria gonorrhoeae. J Clin Microbiol
28. Peters RP, van Agtmael MA, Simoons-Smit AM, Danner SA, Vanden- 2002; 40:3558–64.

Downloaded from https://academic.oup.com/cid/article/45/Supplement_2/S99/285931 by guest on 03 August 2021


broucke-Grauls CM, Savelkoul PH. Rapid identification of pathogens 48. Sandri MT, Lentati P, Benini E, et al. Comparison of the Digene HC2
in blood cultures with a modified fluorescence in situ hybridization assay and the Roche AMPLICOR human papillomavirus (HPV) test
assay. J Clin Microbiol 2006; 44:4186–8. for detection of high-risk HPV genotypes in cervical samples. J Clin
29. Peters RP, Savelkoul PH, Simoons-Smit AM, Danner SA, Vanden- Microbiol 2006; 44:2141–6.
broucke-Grauls CM, van Agtmael MA. Faster identification of path- 49. Hindiyeh M, Hillyard DR, Carroll KC. Evaluation of the Prodesse
ogens in positive blood cultures by fluorescence in situ hybridization Hexaplex multiplex PCR assay for direct detection of seven respiratory
in routine practice. J Clin Microbiol 2006; 44:119–23. viruses in clinical specimens. Am J Clin Pathol 2001; 116:218–24.
30. Wilson DA, Joyce MJ, Hall LS, et al. Multicenter evaluation of a Can- 50. Kehl SC, Henrickson KJ, Hua W, Fan J. Evaluation of the Hexaplex
dida albicans peptide nucleic acid fluorescent in situ hybridization assay for detection of respiratory viruses in children. J Clin Microbiol
probe for characterization of yeast isolates from blood cultures. J Clin 2001; 39:1696–701.
Microbiol 2005; 43:2909–12. 51. Rosey AL, Abachin E, Quesnes G, et al. Development of a broad-range
31. Porcheddu A, Giacomelli G. Peptide nucleic acids (PNAs), a chemical 16S rDNA real-time PCR for the diagnosis of septic arthritis in children.
overview. Curr Med Chem 2005; 12:2561–99. J Microbiol Methods 2007; 68:88–93.
32. Stender H. PNA FISH: an intelligent stain for rapid diagnosis of in- 52. Zucol F, Ammann RA, Berger C, et al. Real-time quantitative broad-
fectious diseases. Expert Rev Mol Diagn 2003; 3:649–55. range PCR assay for detection of the 16S rRNA gene followed by
33. Hayden RT, Isotalo PA, Parrett T, et al. In situ hybridization for the sequencing for species identification. J Clin Microbiol 2006; 44:2750–9.
differentiation of Aspergillus, Fusarium, and Pseudallescheria species in 53. Shrestha NK, Tuohy MJ, Hall GS, Reischl U, Gordon SM, Procop GW.
tissue section. Diagn Mol Pathol 2003; 12:21–6. Detection and differentiation of Mycobacterium tuberculosis and non-
34. Hayden RT, Qian X, Procop GW, Roberts GD, Lloyd RV. In situ hy- tuberculous mycobacterial isolates by real-time PCR. J Clin Microbiol
bridization for the identification of filamentous fungi in tissue section. 2003; 41:5121–6.
Diagn Mol Pathol 2002; 11:119–26. 54. Sandhu GS, Kline BC, Stockman L, Roberts GD. Molecular probes for
35. Hayden RT, Qian X, Roberts GD, Lloyd RV. In situ hybridization for diagnosis of fungal infections. J Clin Microbiol 1995; 33:2913–9.
the identification of yeastlike organisms in tissue section. Diagn Mol 55. Gould SJ. Tempo and mode in the macroevolutionary reconstruction
Pathol 2001; 10:15–23. of Darwinism. Proc Natl Acad Sci U S A 1994; 91:6764–71.
36. Kobayashi M, Urata T, Ikezoe T, et al. Simple detection of the 5S 56. Gould SJ, Eldredge N. Punctuated equilibrium comes of age. Nature
ribosomal RNA of Pneumocystis carinii using in situ hybridisation. J 1993; 366:223–7.
Clin Pathol 1996; 49:712–6. 57. Borst A, Box AT, Fluit AC. False-positive results and contamination
37. Hartmann H, Stender H, Schafer A, Autenrieth IB, Kempf VA. Rapid in nucleic acid amplification assays: suggestions for a prevent and
identification of Staphylococcus aureus in blood cultures by a combi- destroy strategy. Eur J Clin Microbiol Infect Dis 2004; 23:289–99.
nation of fluorescence in situ hybridization using peptide nucleic acid 58. Wilson DA, Yen-Lieberman B, Reischl U, Gordon SM, Procop GW.
probes and flow cytometry. J Clin Microbiol 2005; 43:4855–7. Detection of Legionella pneumophila by real-time PCR for the mip
38. Kempf VA, Mandle T, Schumacher U, Schafer A, Autenrieth IB. Rapid gene. J Clin Microbiol 2003; 41:3327–30.
detection and identification of pathogens in blood cultures by fluo- 59. Farrell JJ, Doyle LJ, Addison RM, Reller LB, Hall GS, Procop GW.
rescence in situ hybridization and flow cytometry. Int J Med Microbiol Broad-range (pan) Salmonella and Salmonella serotype Typhi-specific
2005; 295:47–55. real-time PCR assays: potential tools for the clinical microbiologist.
39. Badak FZ, Goksel S, Sertoz R, et al. Use of nucleic acid probes for Am J Clin Pathol 2005; 123:339–45.
identification of Mycobacterium tuberculosis directly from MB/BacT 60. Odell ID, Cloud JL, Seipp M, Wittwer CT. Rapid species identification
bottles. J Clin Microbiol 1999; 37:1602–5. within the Mycobacterium chelonae-abscessus group by high-resolution
40. Chemaly RF, Tomford JW, Hall GS, Sholtis M, Chua JD, Procop GW. melting analysis of hsp65 PCR products. Am J Clin Pathol 2005; 123:
Rapid diagnosis of Histoplasma capsulatum endocarditis using the 96–101.
AccuProbe on an excised valve. J Clin Microbiol 2001; 39:2640–1. 61. Neoh SH, Brisco MJ, Firgaira FA, Trainor KJ, Turner DR, Morley AA.
41. McNabb A, Adie K, Rodrigues M, Black WA, Isaac-Renton J. Direct Rapid detection of the factor V Leiden (1691 G 1 A) and haemo-
identification of mycobacteria in primary liquid detection media by chromatosis (845 G 1 A) mutation by fluorescence resonance energy
partial sequencing of the 65-kilodalton heat shock protein gene. J Clin transfer (FRET) and real time PCR. J Clin Pathol 1999; 52:766–9.
Microbiol 2006; 44:60–6. 62. Whiley DM, Mackay IM, Sloots TP. Detection and differentiation of
42. Davis TE, Fuller DD. Direct identification of bacterial isolates in blood human polyomaviruses JC and BK by LightCycler PCR. J Clin Micro-
cultures by using a DNA probe. J Clin Microbiol 1991; 29:2193–6. biol 2001; 39:4357–61.
43. Lindholm L, Sarkkinen H. Direct identification of gram-positive cocci 63. Stevenson J, Hymas W, Hillyard D. Effect of sequence polymorphisms
from routine blood cultures by using AccuProbe tests. J Clin Microbiol on performance of two real-time PCR assays for detection of herpes
2004; 42:5609–13. simplex virus. J Clin Microbiol 2005; 43:2391–8.
44. Koliopoulos G, Arbyn M, Martin-Hirsch P, Kyrgiou M, Prendiville W, 64. Levi JE, Kleter B, Quint WG, et al. High prevalence of human pap-
Paraskevaidis E. Diagnostic accuracy of human papillomavirus testing illomavirus (HPV) infections and high frequency of multiple HPV

S110 • CID 2007:45 (Suppl 2) • Procop


genotypes in human immunodeficiency virus-infected women in Bra- sequencing molecular Gram stain, culture, and conventional Gram
zil. J Clin Microbiol 2002; 40:3341–5. stain for diagnosing orthopaedic infections. J Orthop Res 2006; 24:
65. Zekri AR, El-Din HM, Bahnassy AA, et al. TRUGENE sequencing 1641–9.
versus INNO-LiPA for sub-genotyping of HCV genotype-4. J Med Virol 77. Beck RC, Kohn DJ, Tuohy MJ, Prayson RA, Yen-Lieberman B, Procop
2005; 75:412–20. GW. Detection of polyoma virus in brain tissue of patients with pro-
66. Sturmer M, Morgenstern B, Staszewski S, Doerr HW. Evaluation of gressive multifocal leukoencephalopathy by real-time PCR and pyro-
the LiPA HIV-1 RT assay version 1: comparison of sequence and hy- sequencing. Diagn Mol Pathol 2004; 13:15–21.
bridization based genotyping systems. J Clin Virol 2002; 25(Suppl 3): 78. Gharizadeh B, Oggionni M, Zheng B, et al. Type-specific multiple
S65–72. sequencing primers: a novel strategy for reliable and rapid genotyping
67. Suffys PN, da Silva Rocha A, de Oliveira M, et al. Rapid identification of human papillomaviruses by pyrosequencing technology. J Mol Diagn
of Mycobacteria to the species level using INNO-LiPA Mycobacteria, 2005; 7:198–205.
a reverse hybridization assay. J Clin Microbiol 2001; 39:4477–82. 79. Gharizadeh B, Kalantari M, Garcia CA, Johansson B, Nyren P. Typing
68. Wilson DA, Reischl U, Hall GS, Procop GW. Use of partial 16S rRNA of human papillomavirus by pyrosequencing. Lab Invest 2001; 81:
gene sequencing for the identification of Legionella pneumophila and 673–9.

Downloaded from https://academic.oup.com/cid/article/45/Supplement_2/S99/285931 by guest on 03 August 2021


non-pneumophila Legionella spp. J Clin Microbiol 2007; 45:257–8. 80. Chittur SV. DNA microarrays: tools for the 21st Century. Comb Chem
69. Simmon KE, Croft AC, Petti CA. Application of SmartGene IDNS High Throughput Screen 2004; 7:531–7.
software to partial 16S rRNA gene sequences for a diverse group of 81. Mockler TC, Chan S, Sundaresan A, Chen H, Jacobsen SE, Ecker JR.
bacteria in a clinical laboratory. J Clin Microbiol 2006; 44:4400–6. Applications of DNA tiling arrays for whole-genome analysis. Gen-
70. Han XY, Pham AS, Tarrand JJ, Sood PK, Luthra R. Rapid and accurate omics 2005; 85:1–15.
identification of mycobacteria by sequencing hypervariable regions of 82. Brentani RR, Carraro DM, Verjovski-Almeida S, et al. Gene expression
the 16S ribosomal RNA gene. Am J Clin Pathol 2002; 118:796–801. arrays in cancer research: methods and applications. Crit Rev Oncol
71. Hall L, Wohlfiel S, Roberts GD. Experience with the MicroSeq D2 Hematol 2005; 54:95–105.
large-subunit ribosomal DNA sequencing kit for identification of fil- 83. Chen T. DNA microarrays--an armory for combating infectious dis-
amentous fungi encountered in the clinical laboratory. J Clin Microbiol eases in the new century. Infect Disord Drug Targets 2006; 6:263–79.
2004; 42:622–6. 84. Garaizar J, Rementeria A, Porwollik S. DNA microarray technology: a
72. Ronaghi M. Pyrosequencing sheds light on DNA sequencing. Genome new tool for the epidemiological typing of bacterial pathogens? FEMS
Res 2001; 11:3–11. Immunol Med Microbiol 2006; 47:178–89.
73. Diggle MA, Clarke SC. Pyrosequencing: sequence typing at the speed 85. Cebula TA, Jackson SA, Brown EW, Goswami B, LeClerc JE. Chips
of light. Mol Biotechnol 2004; 28:129–37. and SNPs, bugs and thugs: a molecular sleuthing perspective. J Food
74. Tuohy MJ, Hall GS, Sholtis M, Procop GW. Pyrosequencing as a tool Prot 2005; 68:1271–84.
for the identification of common isolates of Mycobacterium sp. Diagn 86. Butcher PD. Microarrays for Mycobacterium tuberculosis. Tuberculosis
Microbiol Infect Dis 2005; 51:245–50. (Edinb) 2004; 84:131–7.
75. Kobayashi N, Bauer TW, Togawa D, et al. A molecular gram stain using 87. Fukushima M, Kakinuma K, Hayashi H, Nagai H, Ito K, Kawaguchi
broad range PCR and pyrosequencing technology: a potentially useful R. Detection and identification of Mycobacterium species isolates by
tool for diagnosing orthopaedic infections. Diagn Mol Pathol 2005; DNA microarray. J Clin Microbiol 2003; 41:2605–15.
14:83–9. 88. Chemlal K, Portaels F. Molecular diagnosis of nontuberculous my-
76. Kobayashi N, Bauer TW, Tuohy MJ, et al. The comparison of pyro- cobacteria. Curr Opin Infect Dis 2003; 16:77–83.

Molecular Diagnostics • CID 2007:45 (Suppl 2) • S111

You might also like