Real-Time PCR in the Microbiology Laboratory

download Real-Time PCR in the Microbiology Laboratory

of 23

Transcript of Real-Time PCR in the Microbiology Laboratory

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    1/23

    R EVIEW 10.1111/j.1198-743X.2004.00722.x

    Real-time PCR in the microbiology laboratoryI. M. Mackay1,2

    1Clinical Virology Research Unit, Sir Albert Sakzewski Virus Research Centre and 2Department of

    Paediatrics, Royal Childrens Hospital, Brisbane, Queensland, Australia

    A B S T R A C T

    Use of PCR in the field of molecular diagnostics has increased to the point where it is now accepted asthe standard method for detecting nucleic acids from a number of sample and microbial types.However, conventional PCR was already an essential tool in the research laboratory. Real-time PCR hascatalysed wider acceptance of PCR because it is more rapid, sensitive and reproducible, while the risk ofcarryover contamination is minimised. There is an increasing number of chemistries which are used todetect PCR products as they accumulate within a closed reaction vessel during real-time PCR. Theseinclude the non-specific DNA-binding fluorophores and the specific, fluorophore-labelled oligonucle-otide probes, some of which will be discussed in detail. It is not only the technology that has changed

    with the introduction of real-time PCR. Accompanying changes have occurred in the traditionalterminology of PCR, and these changes will be highlighted as they occur. Factors that have restricted thedevelopment of multiplex real-time PCR, as well as the role of real-time PCR in the quantitation andgenotyping of the microbial causes of infectious disease, will also be discussed. Because theamplification hardware and the fluorogenic detection chemistries have evolved rapidly, this reviewaims to update the scientist on the current state of the art. Additionally, the advantages, limitations andgeneral background of real-time PCR technology will be reviewed in the context of the microbiologylaboratory.

    Keywords Real-time PCR, quantitation, molecular, diagnostics

    Accepted: 26 December 20021

    Clin Microbiol Infect 2004; 10: 190212

    B A C K G R O U N D

    Diagnostic microbiology is in the midst of a newera. Rapid nucleic acid amplification and detec-tion technologies are quickly displacing the tra-ditional assays based on pathogen phenotyperather than genotype. The polymerase chainreaction (PCR) [1,2] has increasingly been des-cribed as the latest gold standard for detectingsome microbes, but such claims can only be takenseriously when each newly described assay issuitably compared to its characterised predeces-sors [36]. PCR is the most commonly usednucleic acid amplification technique for the

    diagnosis of infectious disease, surpassing theprobe and signal amplification methods. The PCRcan amplify DNA or, when preceded by a reversetranscription (RT) incubation at 4255 C, RNA.RT-PCR is the most sensitive method for thedetection and quantitation of mRNA, especiallyfor low-abundance templates [710]. The PCRprocess can be divided into three steps. First,double-stranded DNA (dsDNA) is separated attemperatures above 90 C. Second, oligonucleo-tide primers generally anneal at 5060 C, and,finally, optimal primer extension occurs at7078 C. The temperature at which the primeranneals is usually referred to as the TM. This is thetemperature at which 50% of the oligonucleotidetarget duplexes have formed. In the case of real-time PCR, the oligonucleotide could represent aprimer or a labelled probe. The TM differs from thedenaturation temperature (TD), which refers to theTM as it applies to the melting of dsDNA. The rate

    Corresponding author and reprint requests: I. M. Mackay,CVRU, SASVRC, Royal Childrens Hospital, Herston Road,Herston, Queensland 4029, AustraliaE-mail: [email protected]

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    2/23

    of temperature change or ramp rate, the length ofthe incubation at each temperature and the num-ber of times each cycle of temperatures is repeatedare controlled by a programmable thermal cycler.Current technologies have significantly shortened

    the ramp rates, and therefore assay time, throughthe use of electronically controlled heating blocksor fan-forced heated air flows.

    The traditional diagnostic microbiologicalassays include microscopy, microbial culture,antigenaemia and serology. These can be limitedby poor sensitivity, slow-growing or poorly viableorganisms, narrow detection windows, complexinterpretation, immunosuppression, antimicrobialtherapy, high levels of background and non-specific cross-reactions [11,12]. Nonetheless,microbial culture produces valuable epidemio-

    logical data, revealing new, uncharacterised oratypical microbes and yielding intact or infectiousorganisms for further study [13]. It is thereforeclear that the role of the traditional assay contin-ues to be an important one [1418]. Additionally,PCR has some significant limitations. Our abilityto design oligonucleotide primers only extends toour knowledge of a microorganisms genome aswell as the ability of publicly available sequencedatabases to suitably represent all variants of thatmicrobe. It is common for microbial genomes tocontain unexpected mutations, which reduce or

    abrogate the function of a PCR. Traditionally,false-positives due to carryover contaminationhave caused considerable problems in the routineimplementation of PCR in the diagnostic laborat-ory and have led to strict guidelines for the designof laboratories dedicated to performing PCR.Additionally, PCR may be too sensitive for someapplications, detecting a microbe that is present atnon-pathogenic levels. Thus, care is requiredwhen designing a PCR assay and interpreting itsresults.

    Existing combinations of PCR and amplicondetection assays will be called conventional PCRthroughout this review. The detection compo-nents include agarose gel electrophoresis [19],Southern blot [20] and ELISA-like systems [21].Conventional PCR has been used to obtain quan-titative data, with promising results [22]. How-ever, these approaches have suffered from thelaborious post-PCR handling steps required toevaluate the amplicon [23].

    The possibility that, in contrast to conventionalPCR, the detection of amplicon could be visual-

    ised as the amplification progressed was awelcome one. This expanded the role of PCRfrom that of a pure research tool to that of aversatile technology permitting the developmentof routine diagnostic applications for the high-

    and low-throughput clinical microbiology labor-atory [24,25]. Along the way, real-time assayshave provided insight into the kinetics of the PCRas well as the efficiency of different nucleic acidextraction methods and the role that some com-pounds play in the inhibition of amplification[20,2633]. Real-time PCR has made many morescientists familiar with the crucial factors contri-buting to successful amplification of nucleic acids.Today, real-time PCR is used to detect nucleicacids from food, vectors used in gene therapyprotocols, genetically modified organisms, and

    areas of human and veterinary microbiology andoncology [3436].The monitoring of accumulating amplicon in

    real time has been made possible by the labellingof primers, oligonucleotide probes (oligoprobes)or amplicons with molecules capable of fluores-cing. These labels produce a change in signalfollowing direct interaction with, or hybridisationto, the amplicon. The signal is related to theamount of amplicon present during each cycleand will increase as the amount of specificamplicon increases. These chemistries have clear

    benefits over earlier radiogenic labels, includingan absence of radioactive emissions, easy disposaland an extended shelf-life [37].

    A significant improvement introduced by real-time PCR is the increased speed with which it canproduce results. This is largely due to the reducedcycle times, removal of separate post-PCR detec-tion procedures, and the use of sensitive fluores-cence detection equipment, allowing earlieramplicon detection [38,39]. A reduced ampliconsize may also play a role in this speed; however, ithas been shown that decreased product size doesnot strictly correlate with improved PCR effi-ciency, and that the distance between the primersand the oligoprobe may play a more significantrole [40,41].

    The technical disadvantages of using real-timePCR instead of conventional PCR include the needto break the seal of an otherwise closed system inorder to monitor amplicon size, the incompatibil-ity of certain platforms with some fluorescentchemistries, and the relatively restricted multiplexcapabilities of current systems. Additionally, the

    Mackay Real-time PCR in the microbiology laboratory 191

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    3/23

    start-up expense of real-time PCR may be prohib-itive for low-throughput laboratories.

    Because most of the popular real-time PCRchemistries involve hybridisation of an oligo-probe(s) to a complementary sequence on one of

    the amplicon strands, the inclusion of more of theprimer that creates that strand is beneficial to thegeneration of an increased fluorescent signal [42].We have found that this asymmetric PCRapproach improves the signal from both ourconventional and real-time oligoprobe-hybridisa-tion assays.

    Although some of the fluorescent labels havebeen given an associated nomenclature by theirdeveloper, the term fluorophore will generally beused to describe these moieties, while their inclu-sion as labels on an oligonucleotide will be

    described as rendering it fluorogenic. The mostcommonly used fluorogenic oligoprobes relyupon fluorescence resonance energy transfer(FRET) between fluorogenic labels or betweenone fluorophore and a dark or black-hole non-fluorescent quencher (NFQ), which dispersesenergy as heat rather than fluorescence [43]. FRETis a spectroscopic process by which energy ispassed between molecules separated by 10100 A

    that have overlapping emission and absorptionspectra [4446]. Forster primarily developed thetheory behind this process, which is a non-radia-

    tive induced dipole interaction [43,47,48].As alluded to earlier, post-amplification mani-pulation of the amplicon is not required for real-time PCR, because the fluorescent signals aredirectly measured as they pass out of the reactionvessel, so real-time PCR is often described as aclosed or homogeneous system. Apart from thetime saved by amplifying and detecting templatein a single tube, there is minimal potential forcarryover contamination, and the assays per-formance can be closely scrutinised withoutintroducing errors due to handling of the ampl-icon [49]. In addition, real-time PCR has proven tobe cost-effective on a per-run basis, when imple-mented in a high-throughput laboratory [50],particularly when replacing conventional, cul-ture-based approaches to microbial detection.

    In the remainder of this review, the theorybehind real-time PCR will be discussed. Addi-tionally, its rapidly expanding use in the study ofhuman infectious disease will provide an exampleof its acceptance and effectiveness in the diagnos-tic microbiology laboratory.

    A M P L I C O N D E T E C T I O N

    It is the detection process that discriminates real-time PCR from conventional PCR assays. There isa range of chemistries currently in use which can

    be broadly categorised as specific or non-specificfor the amplicons sequence [51]. These haverecently been reviewed in detail [52]. Severaladditional reporter systems have since been des-cribed, and these will be discussed below; how-ever, few applications have been described for thespecific detection and genotyping of microbes.

    While the most common oligoprobes are basedon traditional nucleic acid chemistry, the peptidenucleic acid (PNA) is becoming a more popularchoice for oligonucleotide backbones. The PNA isa DNA analogue that is formed of neutral repea-

    ted N-(2-aminoethyl) glycine units instead ofnegatively charged sugar phosphates [53].However, the PNA retains the same sequencerecognition properties as DNA.

    In general, however, the specific and non-specific fluorogenic chemistries detect ampliconwith the same sensitivity [39].

    L I N E A R O L I G O P R O B E S

    The use of a pair of adjacent, fluorogenic hybrid-isation oligoprobes was first described in the late

    1980s [45,54], and, now known as HybProbes,they have become the manufacturers chemistryof choice for the LightCycler (Roche MolecularBiochemicals, Mannheim, Germany), a capillary-based, microvolume fluorimeter and thermocy-cler with rapid temperature control [39,55]. Theupstream oligoprobe is labeled with a 3 donorfluorophore (fluorescein isothiocyanate, FITC),and the downstream probe is commonly labelledwith either a LightCycler Red 640 or Red 705acceptor fluorophore at the 5-terminus, so thatwhen both oligoprobes are hybridised, the twofluorophores are located within 10 nucleotides ofeach other.

    Most recently described fluorogenic oligo-probes fall into the linear class of oligoprobe.The recently described double-stranded oligo-probes function by displacement hybridisation(Fig. 1a) [56]. In this process, a 5 fluorophore-labelled oligonucleotide is, in its resting state,hybridised with a complementary, but shorter,quenching DNA strand that is 3 end-labeled withan NFQ. When the full-length complementary

    192 Clinical Microbiology and Infection, Volume 10 Number 3, March 2004

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    4/23

    Fig. 1. Oligoprobe chemistries. (a) Displacement probes. The shorter NFQ-labeled strand (Q; filled pentagon) is displaced

    when the fluorophore-labelled (F; open circle) strand hybridises to the specific and longer amplicon. (b) Q-PNA primers.Quenching is achieved in the absence of specific template by a short NFQ-labelled PNA molecule designed to hybridisewith the fluorophore-labelled primer. (c) Light-up probes. These PNA probes fluoresce in the presence of a hybridisedDNA strand due to their asymmetric thiazole orange fluorophore (T; open triangle). (d) HyBeacons. In close proximity toDNA, as occurs upon hybridisation with the specific amplicon, the fluorophore emits fluorescence. (e) DzyNA primers.When the primer is duplicated by the complementary strand (dashed line), a DNAzyme is created. In the presence of acomplementary, dual-labelled oligonucleotide substrate, the continuously amplified DNAzyme will specifically cleave thetemplate between the fluorophore and quencher (Q; open pentagon), releasing the labels and allowing fluorescence tooccur. (f) Tripartite molecular beacons. The fluorophore is removed from the NFQs influence upon opening of the hairpinbecause of hybridisation to specific amplicon, permitting fluorescence.

    Mackay Real-time PCR in the microbiology laboratory 193

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    5/23

    sequence in the form of an amplicon is present,the reporter strand will preferentially hybridise tothe longer amplicon, disrupting the quenchedoligoprobe duplex and permitting the fluoro-phore to emit its excitation energy directly.

    This technique can also be used with a fluor-ophore-labelled primer, and, due to the addedstringency of the complementary strand, thesystem acts as its own hot-start, as was shownusing an NFQ-labeled PNA [57] strand (Q-PNA)(Fig. 1b) [58]. In this system, the quenching probeis bound to unincorporated fluorogenic primersuch that the NFQ and fluorophore are adjacent,resulting in a quenched system. Once the dsDNAamplicon is created by primer extension, how-ever, the Q-PNA is displaced, and the fluorophorecan fluoresce. The PNA backbones cannot be

    extended or hydrolysed by a DNA polymerase.The light-up probe is also a PNA to which theasymmetric cyanine fluorophore thiazole orangeis attached (Fig. 1c) [59]. When hybridised with anucleic acid target, as either a duplex or triplex,the fluorophore becomes strongly fluorescent.These oligoprobes do not interfere with the PCRor require conformational change, they are sensi-tive to single nucleotide mismatches and, becausea single reporter is used, they allow the directmeasurement of fluorescence instead of the meas-urement of a change in fluorescence between two

    fluorophores [59,60]. However, non-specific fluor-escence has been reported during extended cyc-ling [61].

    The HyBeacon is a single linear oligonucleotideinternally labelled with a fluorophore that emitsan increased signal upon formation of a duplexwith the target DNA strand (Fig. 1d) [62,63]. TheHyBeacon is labelled at the 3-terminus with aphosphate or octanediol molecule to prevent Taq-mediated extension. This technique is used withall the non-incorporating nucleotide-based oligo-probe chemistries used in real-time PCR to ensurethat they do not function as a primer. Thischemistry does not require destruction, interac-tion with a second oligoprobe or secondarystructure changes to produce a signal, and it isrelatively cheap and simple to design.

    D U A L - L A B E L L E D O L I G O P R O B E S

    In the early 1990s, an innovative approachinvolved nick-translation PCR in combinationwith dual-fluorophore-labelled oligoprobes was

    introduced [26]. In the first truly homogeneousassay of its kind, a fluorophore was added to the5-terminus and another to the middle of asequence-specific oligoprobe. When in such closeproximity, the 5 reporter fluorophore (6-carboxy-

    fluoroscein; FAM) transferred laser-inducedexcitation energy by FRET to the 3 quencherfluorophore (6-carboxy-tetramethyl-rhodamine;TAMRA). The oligoprobe hybridised to its tem-plate prior to the extension step, and the fluor-ophores were subsequently released during theprimer extension step as a result of the 5 to 3endonuclease activity of a suitable DNA polym-erase. Once the labels were separated, the repor-ters emissions were no longer quenched, and theinstrument monitored the resulting fluorescence.Today, these oligoprobes are labelled at each

    terminus and are called 5

    nuclease, hydrolysis orTaqMan oligoprobes. The nuclease oligoprobe isthe manufacturers chemistry of choice for theABI Prism sequence detection systems.

    A modification of the 5 nuclease chemistry hasresulted in the minor groove binding (MGB)oligoprobes [64]. This chemistry, commerciallycalled the Eclipse oligoprobes, replaces the Taq-Man oligoprobes standard TAMRA quencherwith a proprietary NFQ and incorporates amolecule that hyperstabilises the oligoprobetar-get duplex by folding into the minor groove of the

    dsDNA [65,66]. A fluorophore is attached to the3 end, and in the unbound state the oligoprobeassumes a random coil configuration that isefficiently quenched. This chemistry allows theuse of very short (1217-nucleotide) oligoprobesbecause of a 1530 C rise in their TM resultingfrom the interaction of the MGB with the DNAhelix. These short oligoprobes are ideal for detect-ing single-nucleotide polymorphisms (SNPs),because they are more significantly destabilisedby nucleotide changes within the hybridisationsite than are larger oligoprobes.

    Another dual-labelled oligonucleotide sequ-ence has been used as the signal-generatingportion of the DzyNA-PCR system (Fig. 1e) [67].Here, the reporter and quencher molecules areseparated following specific cleavage of the oligo-nucleotides holding them in close proximity. Thiscleavage is performed by a DNAzyme, which iscreated during the PCR as the complement ofan antisense DNAzyme sequence included inthe 5 tail of one of the primers. Upon clea-vage, the fluorophores are released, allowing

    194 Clinical Microbiology and Infection, Volume 10 Number 3, March 2004

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    6/23

    the production of fluorescence in an identicalmanner to a hydrolysed TaqMan oligoprobe.

    H A I R P I N O L I G O N U C L E O T I D E S

    Molecular beacons were the first hairpin oligo-probes to be used in real-time PCR. The molecularbeacons fluorogenic labels are positioned at thetermini of the oligoprobe. The labels are held inclose proximity by distal stem regions of homol-ogous base pairing deliberately designed to createa hairpin structure. The closed hairpin isquenched due either to FRET or direct collisiontransfer of energy occurring at the molecular levelas a consequence of the intimate proximity of thelabels [68]. In the presence of a complementarysequence, designed to occur within the bounds of

    the primer binding sites, the oligoprobe willhybridise, shifting into an open configuration.The fluorophore is now spatially removed fromthe quenchers influence, allowing fluorescentemissions to be monitored [69]. This structuralchange occurs in each cycle, increasing in cumu-lative intensity as the amount of specific ampliconincreases. The quencher, DABCYL (4-(4-dimeth-ylamino-phenylazo)-benzene), differs from thatdescribed for the nuclease oligoprobes because itis an NFQ.

    Recently, tripartite molecular beacons have

    been added to this class of fluorogenic chemistry(Fig. 1f) [70]. These oligoprobes have beendesigned to fulfill a need for suitably high-throughput chemistries and they combine amolecular beacons hairpin with long or unla-belled single-stranded arms, each designed tohybridise to an oligonucleotide labelled witheither a fluorophore or an NFQ. The system isquenched in the hairpin state due to the closeproximity of the labels, but fluorescent whenhybridised to the specific amplicon strand.Because the function of these oligoprobes dependsupon correct hybridisation of the stem and twooligoprobes, their accurate design is crucial [8].

    Finally, a self-quenching hairpin primer hasrecently been described which is commerciallyentitled the light upon extension (LUX) fluoro-genic primer [71]. This chemistry is dark in theabsence of specific amplicon, through the naturalquenching ability of a carefully placed guanosinenucleotide. The natural quencher is broughtinto close proximity with the FAM or JOE5 2,7-dimethoxy-4,5-dichloro-6-carboxy-fluoroscein

    fluorophore via a stretch of 5 and 3 comple-mentary sequences. In the presence of specifictarget, the primer hybridises, opening the hairpinand permitting fluorescence from the fluoro-phore.

    M I C R O B I A L Q U A N T I T A T I O N

    Although the terminology is often confused, real-time PCR does not inherently imply quantitativePCR. To quantify the amount of template presentin a sample, thought must be given to the typeand number of controls required. Standards areused to allow calculation of the amount oftemplate present in a patient sample, whileinternal controls (ICs) are mostly used to deter-mine the occurrence of false-negative reactions,

    examine the ability to amplify from a preparationof nucleic acids, and, more rarely in real-timePCR, as a standard for quantitation. Certainly, thereliability of quantitative PCR methods is inti-mately associated with the choice and quality ofthe assay controls [72,73].

    No matter what control is chosen, it is imper-ative to accurately determine its concentration[74] and to ensure that ICs are added at suitablelevels in order to prevent extreme competitionwith the wild-type template for reagents [75]. Theuse of a spectrometer is inadequate for quantitat-

    ing a control molecule [76]; however, in combi-nation with an experimental and statisticalanalysis, the reliability of the values is greatlyenhanced [7781]. Finally, one must rememberthat the results of quantitation using a molecularcontrol need to be expressed relative to a suitablebiological marker, e.g., in terms of the volume ofplasma, the number of cells or the mass of tissueor genomic nucleic acid, thus allowing compar-ability between assay results and testing sites [82].

    Standards for quantitation

    Most commonly, an exogenous control is createdusing a cloned amplicon, a portion of the targetorganisms genome, or simply the purified ampl-icon itself [83]. This control forms the basis of anexternal standard curve created from the dataproduced by the individual amplification of adilution series of exogenous control. The concen-tration of an unknown, which is amplified in thesame reaction, but in a separate vessel, can thenbe found from the standard curve. While the

    Mackay Real-time PCR in the microbiology laboratory 195

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    7/23

    external standard curve is the more commonlydescribed quantitative approach, it frequentlysuffers from uncontrolled and unmonitored inter-vessel variations. Some platforms have overcomethis issue by including a capacity to detect and

    correct for variation in the emissions of a non-participating, or passive, internal referencefluorophore (6-carboxy-N,N,N,N-tetramethylrhod-amine; ROX). The corrected values, obtained froma ratio of the emission intensity of the fluorophoreand ROX, are called RQ+. To further controlamplification fluctuations, the fluorescence from ano-template control reaction (RQ) is subtractedfrom RQ+, resulting in the DRQ value thatindicates the magnitude of the signal generatedfor the given PCR [84]. Assays that lack thiscapacity are more appropriately described as

    semiquantitative.

    Internal controls (ICs)

    The use of an IC was described in the earliest ofPCR experiments as an important quality con-trol [85,86], particularly when performing com-petitive quantitation. When such a control isadded before template purification (extractioncontrol) or amplification (amplification control),it is called an exogenous IC, since it does notoccur naturally within the nucleic acid pre-

    paration, but is co-amplified within the samereaction. Ideally, the IC should hybridise to thesame primers, have an identical amplificationefficiency [74,87], and contain a discrimin-ating feature such as a change in its length[72,72,75,88,89] or, more commonly in todaysoligoprobe-based methods, a change in the seq-uence [73,90] of the wild-type target [91,92].However, IC templates that bind different prim-ers or have different amplification efficienciescan still prove useful as standards for semi-quantitative PCR or relative quantitation.

    An endogenous control is a template thatoccurs naturally within the specimen being exam-ined. Housekeeping genes often fulfill this role,and they have been successfully used to quanti-tate gene expression by RT-PCR and monitor theintegrity of a template after its purification [85].When endogenous controls are used for thequantitation of RNA, it is essential that thehousekeeping gene is minimally regulated andexhibits a constant and cell cycle-independentbasal level of transcription [93]. This is not the

    case for some commonly used genes such asb-actin, whereas studies have shown that an 18SrRNA target meets the desired criteria [93,94].

    Relative vs. absolute quantitation

    The amount of template in a sample can bedescribed either relatively or absolutely. Relativequantitation is the simpler approach, and des-cribes changes in the amount of a target sequencecompared to its level in a related matrix or withinthe same matrix by comparison to the signal froman endogenous or other reference control. Abso-lute quantitation is more demanding but statesthe exact number of nucleic acid targets present inthe sample in relation to a specific unit, making iteasier to compare data from different assays and

    laboratories [7,95]. Absolute quantitation may benecessary when there is a lack of sequentialspecimens to demonstrate a relative change inmicrobial load, or when no suitably standardisedreference reagent is available.

    A highly accurate approach used for absolutequantitation by conventional PCR utilises com-petitive coamplification of one or a series of ICs ofknown concentration with a wild-type targetnucleic acid of unknown concentration [9699].However, conventional competitive quantitationis technically demanding, requiring significant

    development and optimisation compared toquantitation by real-time PCR, which is bettersuited to the quick decision-making required in aclinical environment [100102]. Software with theability to calculate the concentration of anunknown by comparing real-time PCR signalsgenerated by a coamplified target and IC is rarebut emerging [7]. In addition, new or improvedformulae are appearing which aim to makequantitation more reliable and simpler [103].

    Acquisition of fluorescence data

    Fluorescence data generated by real-time PCRassays are generally collected from PCR cyclesthat occur within the linear amplification portionof the reaction, where conditions are optimal andthe fluorescence accumulates in proportion to theamplicon [52] (Fig. 2). This is in contrast to signaldetection from the endpoint of the reaction, wherethe final amount of amplicon may have beenaffected by inhibitors, poorly optimised reactionconditions or saturation effects due to the

    196 Clinical Microbiology and Infection, Volume 10 Number 3, March 2004

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    8/23

    presence of excess double-stranded amplicon. Infact, at the endpoint there may be no relationshipbetween the initial template and final ampliconconcentrations. Because the emissions from fluor-escent chemistries are temperature-dependent,data are generally acquired only oncecycle, atthe same temperature [55].

    The fractional cycle number at which the real-time fluorescence signal mirrors progression ofthe reaction above the background noise is usedas an indicator of successful target amplification[104]. Most commonly, this is called the thresholdcycle (CT), but a similar value is described for theLightCycler, and the fractional cycle is called thecrossing point (CP). The CT is defined as the PCR

    cycle in which the gain in fluorescence generatedby the accumulating amplicon exceeds ten stand-ard deviations of the mean baseline fluorescence,using data taken from cycles 315 [105]. The CTand CP are proportional to the number of target

    copies present in the sample [29] and are assumedto represent equal amounts of amplicon present ineach tube or capillary, since the CT and CP valuesrepresent the fractional cycle number for eachsample at a single fluorescence intensity value. Inpractice, the CT and CP are calculated after thedefinition of a noise band that excludes data fromearly PCR cycles that cannot be distinguishedfrom background noise. The final CT and CPvalues are the fractional cycles at which a singlefluorescence value (usually at or close to the noiseband) intersects each samples plotted PCR curve

    [104] (Fig. 2). The accuracy of the CT or CPdepends upon the concentration and nature of thefluorescence-generating component, the amountof template initially present, the sensitivity of theplatform, and the platforms ability to discrimin-ate specific fluorescence from background noise.

    Improved quantitation using real-time PCR

    Significant improvements in the quantitation ofmicrobial load by real-time PCR result from thedetection systems enormous dynamic range,

    which can accommodate at least eight log10

    copiesof nucleic acid template [92,100,106114]. Thebroad dynamic range avoids the need for pre-dilution of an amplicon before detection, or theneed to repeat an assay using a diluted samplebecause a preliminary result falls outside thelimits of the assay. Both of these problems occurcommonly when using conventional endpointPCR assays for quantitation, as their detectionsystems are unable to encompass the products ofhigh template loads while maintaining adequatesensitivity [113,115117]. The flexibility of real-time PCR is further demonstrated by its ability todetect one target in the presence of a vast excessof another target during duplexed assays [109].

    Real-time PCR is also a particularly attractivealternative to conventional PCR for the study ofmicrobial load because of its low inter-assay andintra-assay variability [100,112,118] and its equiv-alent or improved sensitivity compared to micro-bial culture, or conventional single-round andnested PCR [17,100,110,119126]. Real-time PCRhas been reported to be at least as sensitive as

    Fig. 2. Kinetic analysis. The ideal amplification curve of areal-time PCR (solid), when plotted as fluorescence inten-sity against the cycle number, is a sigmoidal curve. Earlyamplification cannot be viewed because the emissions aremasked by the background noise. However, when enoughamplicon is present, the assays exponential progress canbe monitored as the rate of amplification enters a linearphase (LP). Under ideal conditions, the amount of ampl-icon increases at a rate of one log10 every 3.32 cycles. Asprimers and enzyme become limiting, and productsinhibitory to the PCR and overly competitive to oligoprobehybridisation accumulate, the reaction slows, entering atransition phase (TP) and eventually reaching a plateauphase (PP) where there is little or no increase in fluores-cence. The point at which the fluorescence surpasses thenoise threshold (dashed horizontal line) is called the

    threshold cycle or crossing point (CT

    or CP

    ; indicated byan arrow), and this value is used in the calculation oftemplate quantity during quantitative real-time PCR. Alsoshown are curves representing a titration of template(dashed curves), consisting of decreasing starting templateconcentrations, which produce higher CT or CP values,respectively. Data for the construction of a standard curveare taken from the LP.

    Mackay Real-time PCR in the microbiology laboratory 197

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    9/23

    Southern blot, still considered by some as the goldstandard for probe-based hybridisation assays[122].

    M I C R O B I A L G E N O T Y P I N G

    Although nucleotide sequencing is still the goldstandard for characterising unknown nucleicacids, it is a relatively lengthy process. Thedevelopment of real-time PCR has partiallyaddressed this failing by providing a tool capableof routine detection of characterised mutations,insertions or deletions.

    Most fluorescent chemistries used for real-timePCR do not rely upon a destructive process togenerate a signal. Therefore, they may be able toperform a genotyping role at the completion of

    the PCR. The SYBR green and HybProbe chem-istries are most commonly used to perform theseanalyses; however, the double-stranded and light-up oligoprobes and HyBeacons should also func-tion in this role. Other chemistries, such as theTaqMan and Eclipse oligoprobes and hairpinoligonucleotides, discriminate these nucleotidechanges using two sets of oligoprobes to differ-entiate the wild-type from the altered sequences.While this is a perfectly legitimate and functionalapproach to genotyping by real-time PCR, theextra fluorogenic oligonucleotides increase the

    overall cost of the assay. Additionally, the num-ber of different microbes that can be discrimin-ated during multiplex real-time PCR is reduced,since two fluorophores must be assigned toanalyse each microbe. The occurrence of a mis-match between a hairpin oligonucleotide and itstarget has a greater destabilising effect on theduplex than the introduction of an equivalentmismatch between the target and a linear oligo-probe. This is because the hairpin structureprovides a highly stable alternative configuration.Therefore, hairpin oligonucleotides are more spe-cific than the more common linear oligoprobes,making them ideal candidates for detecting SNPs[68].

    Genotyping data are obtained after the com-pletion of the PCR, and therefore represent anendpoint analysis. The amplicon is denatured andrapidly cooled to encourage the formation offluorophore and target strand complexes. Thetemperature is then gradually raised, and thefluorescence from each vessel is continuouslyrecorded. The detection of sequence variation

    using fluorescent chemistries relies upon thedestabilisation incurred as a result of the chan-ge(s). The non-specific chemistries reflect thesechanges in the context of the entire dsDNAamplicon, requiring the dissociation of fluorogen-

    ic molecules from the dsDNA, which only occursupon melting of the duplex. The sequence chan-ges have a different impact upon the specificfluorogenic chemistries, altering the expected TMin a manner that reflects the particular nucleotidechange. The resulting rapid decrease in fluores-cence using either approach can be presented as amelt peak using software capable of calculatingthe negative derivative of the fluorescence changewith temperature (Fig. 3).

    Importantly, different nucleotide changesdestabilise hybridisation to different degrees,

    and this can be incorporated into the design ofgenotyping assays to ensure maximum discrim-ination between melt peaks. The least destabilis-ing mismatches include G (G:T, G:A and G:G),whereas the most destabilising include C (C:C,C:A and C:T) [127].

    M U L T I P L E X R E A L - T I M E P C R

    Multiplex PCR uses one or more primer sets topotentially amplify multiple templates within asingle reaction [128,129]. However, its use in real-

    time PCR has led to confusion in the traditionalterminology. Multiplex real-time PCR more com-monly refers to the use of multiple fluorogenicoligoprobes for the discrimination of ampliconsthat may have been produced by one or severalprimer pairs. The development of multiplex real-time PCR has proven problematic because of thelimited number of fluorophores available [26] andthe frequent use of monochromatic energisinglight sources. Although excitation by a singlewavelength produces bright emissions from asuitably receptive fluorophore, the number offluorophores that can be excited by that wave-length is limited [130].

    The discovery and application of the non-fluorescent quenchers has made available somewavelengths that were previously occupied bythe emissions from the early quenchers them-selves. This should permit the future inclusion ofa greater number of spectrally discernable oligo-probes/reaction, and highlights the need for asingle non-fluorescent quencher that can quencha broad range of emission wavelengths (e.g.,

    198 Clinical Microbiology and Infection, Volume 10 Number 3, March 2004

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    10/23

    400600 nm). The impressive electron-donatingproperties of guanosine make it an ideal naturalquencher, and its use has contributed to thegrowing number of assays that only require asingle fluorophoretarget [131].

    Early real-time PCR systems contained optim-ised filter sets to minimise overlap of the emissionspectra from the fluorophores. Despite this, thenumber of fluorophores that could be combinedand clearly distinguished was limited. Morerecent real-time PCR platforms have incorporatedeither multiple light-emitting diodes, or a tung-sten light source that emits over a wide range ofwavelengths. When these platforms also incor-

    porate high-quality optical filters, it is possible touse many of the current real-time PCR detectionchemistries on the one machine. Unfortunatelysome platforms are not suitably constructed. Evenif these improvements are included, the platform

    can still only perform four-colour oligoprobemultiplexing, and one colour is ideally set asidefor use as an IC. Some real-time PCR designs havemade use of conserved single or multiple nucleo-tide changes among similar templates to allowtheir differentiation by concurrent changes to theoligoprobes TM or the amplicons TD [132,133].Combining the use of multiple fluorophores withthe discrimination of additional targets by tem-perature allows the identification of a significantlylarger number of amplicon targets [134]; however,this combined approach has not been applied to

    the diagnosis of infectious disease on a significantscale [135], possibly because of the sequencevariation among many microbial genes [119,136139]. Far more commonly, this approach has beenused for the detection of human genetic diseases,where as many as 27 possible nucleotide substi-tutions have been detected using only one or twofluorophores [140147].

    To date, there have been only a handful ofdiagnostic microbial assays that can trulyco-amplify and discriminate more than two fluor-ophores. An impressive multiplex, real-time PCR

    protocol discriminated between four retroviraltarget sequences [148]; however, conventionalmultiplex PCR using endpoint detection haseasily discriminated between more than fivedifferent amplified sequences, indicating a greaterdegree of flexibility [149154].

    Future development of novel chemistries andimproved real-time instrumentation and softwareshould significantly improve the ability to multi-plex fluorophores for enhanced real-time PCRassays. Perhaps a chimera of real-time PCR andmicroarray technology, in combination withmicrofluidic devices, may advance all three tech-nologies to a point where the desired number oftemplates could be easily amplified and discrim-inated.

    S P E C I F I C A P P L I C A T I O N S F O RM I C R O B I O L O G Y

    Real-time PCR assays have been extremely usefulfor studying microbial agents of infectious dis-ease, where they have helped to clarify many

    Fig. 3. Fluorescence melting curve analysis. At the com-pletion of a real-time PCR using a fluorogenic chemistry,the reaction can be cooled to a temperature below theexpected TM of the oligoprobes and then heated to above90C at a fraction of a degree/second (a). During heating,the emissions of the reporter or acceptor fluorophore canbe constantly acquired (b). Software calculates the negativederivative of the fluorescence with temperature, producinga clear melt peak that indicates the TM of the oligoprobetarget melting transition (black peak; c) or the TD ofmelting dsDNA. When one or more nucleotide changes arepresent, the TM or TD is shifted (grey peak). This shift isreproducible and can be used diagnostically to genotypemicrobial templates.

    Mackay Real-time PCR in the microbiology laboratory 199

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    11/23

    disease processes. Most of the assays presented inthe literature have increased the frequency ofmicrobial detection compared to non-PCR tech-niques, making the implementation of real-timePCR attractive to many.

    Of course, real-time PCR has also provenvaluable for basic microbiological research, whereits ability to amplify template from a wide arrayof sample types (Table 1) has made it an idealsystem for application across the various micro-biological disciplines [155]. Increasingly, theseapplications are difficult to review, due to theiruse as a tool within, rather than the focus of, apublished study.

    Viruses

    Within microbiology, the application of real-timePCR has had the biggest impact upon the field ofvirology, where studies have qualitatively inves-tigated the role of viruses in a range of humandiseases [156]. Also, epidemiological studies ofco-infections have been improved by thesemolecular techniques, which can reliably measurethe amount of two nucleic acid targets presentwithin a single sample [119,157,158]. Real-timePCR has also improved the discrimination ofmultiple viral genotypes within a single reactionvessel [159] and provided an alternative to mor-

    bidity and mortality assays for virus detection. Anexample is Newcastle disease virus, which existsas two radically different pathogenic phenotypescaused by small nucleotide changes that can be

    easily detected using fluorescence melting curveanalysis to reveal the genetic pathotype of thestrain [160].

    Direct and indirect links between viral infectionand chronic conditions such as sarcoma [121,161

    164], carcinoma [122,165], cervical intra-epithelialneoplasia [166168] and lymphoproliferativedisorders [169,170] can be relatively easilystudied using real-time PCR. Other studies havedescribed the presence of flaviviruses [106,126,171176], hepadnaviruses [113,115,177], herpesvi-ruses [30,40,100,102,107109,116,119,121,122,137,155,158,165,178187], orthomyxoviruses [125],parvoviruses [92], papovaviruses [32,139,159],paramyxoviruses [124,160,188], pestiviruses [189],picornaviruses [110,111,190195], poxviruses [196],retroviruses [118,123,197200], rhabdoviruses

    [201] and TT virus [202].A significant number of studies have used PCRto detect viral load, and have proved its useful-ness as an indicator of the extent of activeinfection, interactions between virus and host,and the changes in viral load as a result ofantiviral therapies, all of which can play a role inthe treatment regimen selected [203205]. Con-ventional quantitative PCR has already proventhat the application of nucleic acid amplificationto the monitoring of viral load provides a usefulmarker of disease progression and the efficacy of

    antiviral compounds [97,204,206210]. Becausedisease severity and viral load are linked, theuse of real-time PCR quantitation has provenbeneficial when studying the role of viral reacti-vation or persistence in the progression of disease[40,102,107,108,119,158,165,171,183,184,187,211,211216]. Alterations to a microbes tropism or itsreplication, and the effects that these changeshave on a host cell, can also be followed usingreal-time PCR [217219].

    The role of highly sensitive and rapid real-timePCR assays in the thorough assessment of viralgene therapy vectors before their use in clinicaltrials has become an important one. Nucleaseoligoprobes have been most commonly used forthese studies, which assess the biodistribution,function and purity of the novel drug prepara-tions [199,220225].

    Likewise, the study of new and emergingviruses has been ideally complemented by theuse of homogeneous real-time PCR assays as toolsto demonstrate and strengthen epidemiologicallinks between unique viral sequences and the

    Table 1. An incomplete list indicating the extraordinaryvariety of sample types from which nucleic acids can besuccessfully prepared, amplified and detected using real-time PCR assays

    Nucleic acid origins References

    Plants [293]Animals [111,260]Urban sludge [110]Microbial culture [177,234,240,253,254,266,267,

    280,284,292,294]Solid tissues [172,198,238,256258,271,288,295]

    Cerebrospinal fluid [136,190,192,194,248]Peripheral blood

    mononuclear cells[183]

    Bone marrow [120]Whole blood [179,291]Plasma [118]Serum [171,172,180,279,287]Swabs [192,259,296]Bronchoalveolar lavage [243,246]Amniotic fluid [286]Saliva and sputum [158,233]Faeces [264,285]Urine [12,155,178]

    200 Clinical Microbiology and Infection, Volume 10 Number 3, March 2004

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    12/23

    clinical signs and symptoms experienced bypatients [124,126,201,226230].

    The speed and flexibility of real-time PCR hasalso proven useful for commercial interests whorequire exquisite sensitivity to screen for micro-

    bial contamination within large-scale reagentpreparations produced from eukaryotic expres-sion systems [231,232].

    Bacteria

    The benefits to the patient from rapid real-timePCR assays are most notable when applied to thedetection of bacteria. The results can quicklyinform the clinician as to the infection status ofthe patient, allowing a more specific and timelyapplication of antibiotics. This can limit the

    potential for toxicity due to shotgun treatmentregimens, reduce the duration of a hospital stayand prevent the improper use of antibiotics, thusminimising the potential for resistant strains toemerge.

    Broad applications of real-time PCR can aug-ment or replace traditional culture or histochem-ical assays, as was seen with the creation of amolecular assay capable of classifying bacteria inthe same way as a Gram stain [233]. However,specific bacterial species are more frequently thefocus for real-time PCR assays, especially when

    long culture times can be replaced by rapid andspecific gene detection. Leptospira genospecies,Mycobacterium and Propionibacterium spp., Chla-mydia spp., Legionella pneumophila and Listeriamonocytogenes have all been detected and in somecases quantitated with the use of real-time PCRassays [41,234246].

    The detection of Neisseria gonorrhoeae has bene-fited from real-time PCR, particularly in the roleof a confirmatory test when the specificity ofcommercial assays fails [247]. This example high-lights the need for care when choosing a bacterialPCR target, especially when that target exists on aplasmid that is exchanged among other bacteria,providing potentially confusing diagnosticresults. Neisseria meningitidis causes meningococ-cal disease, and real-time PCR has proven to be apowerful tool that can be quickly developed forthe rapid discrimination of currently circulatingpathogens [248].

    The detection and monitoring of antibioticresistance among clinical isolates ofStaphylococcusaureus, Staphylococcus epidermidis, Helicobacter

    pylori, Enterococcus faecalis and Enterococcus fae-cium has also benefited from real-time applica-tions [249258]. Additionally, the understandingand treatment of fulminant diseases such asmeningitis, sepsis, inflammatory bowel disease

    and food-poisoning caused by characterised bac-teria such as the group B streptococci and Myco-bacterium spp., Escherichia coli and Bacteroidesvulgatus [259265] have been enhanced by thespeedy return of results, which also aids trackingof microbial outbreaks to their source.

    Real-time PCR has made possible the rapidquantitation and differentiation of some of themore exotic pathogenic bacteria, such as the tick-borne spirochete Borrelia burgdorferi [266268] andthe methanotropic bioremediating Methylocystisspp. [269].

    The involvement of treponemes in the devel-opment of periodontal disease has been studiedusing 5 nuclease chemistry, revealing a microbialrole in every stage [270]. In addition, measure-ment of the bacterial load of Tropheryma whippleihas allowed the discrimination of environmentalcontamination and low-level colonisation fromactive infection [271].

    More recently, there has been an explosion ofliterature indicating that real-time PCR is the toolof choice for the rapid detection of microbes usedas agents of biological warfare. In some cases, the

    assays have allowed rapid discrimination ofweaponised pathogens from the harmless labor-atory-adapted or vaccine-related strains. At theforefront of the available literature are assays todetect Bacillus anthracis spores and the bacteriumsvirulence-encoding plasmids or chromosomalmarkers [272,273,273277]. Conventional assaysmay take 48 h to complete, and, for obviousreasons, this is an unacceptable lag period.

    Fungi, parasites and protozoans

    The smallest number of applications have beenrelated to the study of fungal, parasitic andprotozoan pathogens of humans. Nonetheless,real-time PCR assays have significantly contribu-ted to the general diagnosis of invasive diseasecaused by Aspergillus fumigatus and Aspergillus

    flavus [278,279]. Also, monitoring the transcriptionlevels of certain Aspergillus nidulans transportergenes has provided important information abouttheir role in multiresistance [280]. In addition,real-time assays have been used when investi-

    Mackay Real-time PCR in the microbiology laboratory 201

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    13/23

    gating buildings for the presence of potentiallyharmful levels of toxigenic fungal spores, orconidia, such as those produced by Stachybotryschartarum [281283].

    Cryptosporidium parvum oocysts and the spores

    from Encephalitozoon spp. have been successfullygenotyped or speciated using real-time PCR,which has significantly improved on laboratorydiagnosis using microscopy and histochemicalstaining, especially for low concentrations ofexcreted material [284,285].

    Rapid serological detection ofToxoplasma gondiiis often hampered by the presence of the parasitein patients who are immunocompromised. Addi-tionally, the length of time required for traditionalculture or mouse inoculation is excessive. There-fore, rapid molecular methods have vastly

    improved the detection of this microbe [120,286].Additionally, this technology is useful for thestudy of T. gondii responses to antimicrobialtherapies [287].

    Detection of malarial parasites using a mousemodel in combination with real-time PCR hasimproved result turnaround time and meant thatparasite load data can be generated [288,289].Real-time PCR has also proven useful for directin-vivo detection and quantitation of malarialparasites with a high level of sensitivity [290,291],in addition to monitoring the stage-specific mat-

    uration ofPlasmodium falciparum via the transcrip-tion of specific genes [292].

    C O N C L U S I O N S A N D S U M M A R Y

    Microbiology is ideally suited for the benefits ofsensitivity and rapidity that PCR has brought tothe research laboratory. However, the advent ofreal-time PCR has further improved the role ofPCR in the high-throughput environment byadding a detection system capable of enormousdynamic range, homogeneity of amplification anddetection, and the ability to genotype an ampli-fied nucleic acid without the need for additionalsteps. Unfortunately, many of the genotypingapplications have been trialled first in the field ofhuman genetics, where there is frequently a moreabundant source of template, and the geneticchanges, once characterised, remain constant.Nonetheless, this review has highlighted thegeneral acceptance of real-time PCR in theresearch and diagnostic microbiology laboratory,and its popularity is continuing to expand.

    Advances in the development of fluorophores,nucleotide labelling and the novel application ofoligoprobe hybridisation have provided real-timePCR technology with a broad enough commercialbase to promote its usefulness to the wider non-

    research scientific community. Robotic nucleicacid extraction and liquid-handling systems, com-bined with rapid thermal cyclers and instrumen-tation capable of detecting and differentiatingmultiple amplicons using many of the chemistriesdescribed in this and other reviews, make real-time PCR an attractive and viable proposition forthe routine diagnostic laboratory. Many laborat-ories rely upon tissue culture to isolate microbialagents of infectious disease, in combination withserological methods to further confirm the iden-tity of the isolates or to monitor a patients

    immune response to an infectious agent. Suchmethods, while providing an important source ofinformation about unknown and emerging path-ogens, may take a prolonged and clinically signi-ficant amount of time to complete.

    According to the literature, the most widelyused fluorogenic probe format is the 5 nucleaseoligoprobe, although that is most likely due to itscommercial maturity. The rate of publicationsdescribing other methods, especially those utilis-ing the LightCycler in combination with a pair ofHybProbes, is significant and changing the bal-

    ance rapidly, especially in the area of microbialdetection and genotyping. There are also morevirus-detecting real-time PCR applications des-cribed in the literature than for any other microbe.The more recently developed oligoprobe chemis-tries have been used in only a few innovativeapplications, but they will be better understood astheir benefits and limitations are more widelydescribed, and hopefully they will allow a greatervariety of options for microbial genotyping.

    Recent developments in multiplex real-timePCR have suggested a future in which easyidentification, genotyping and quantitation ofmicrobial targets in single, rapid reactions willbe commonplace. Of course, real-time PCR is byno means restricted to microbiology, as significantachievements have already been made in the areaof human genetic diagnostics, applying all thebenefits of real-time PCR to enhance the detectionof genetic disease. However, the technology isonly as reliable as the accompanying controls andassociated quality assurance programmes. Thisincludes the quality of standards, the use of

    202 Clinical Microbiology and Infection, Volume 10 Number 3, March 2004

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    14/23

    suitably controlled standard curves, and the needto fully optimise, validate and evaluate each andevery new assay against previously standardisedassays. Without such care, real-time PCR willprovide fast but inaccurate data to the clinician,

    who will surely come to rely upon such assays,as they represent a growing proportion of theresult-generating tests within the diagnosticmicrobiology environment. In addition, commer-cial interests will undoubtedly play an expandingrole in determining which technologies enter intothe mainstream.

    Perhaps in combination with micro- andmacroarray technology and emerging microflui-dic devices, real-time PCR assays that can dis-criminate as many targets as desired, whileproducing quantitative data at a greatly increased

    speed, will consolidate fluorogenic nucleic acidamplification as a routine and incredibly power-ful tool for the laboratory of tomorrow.

    A C K N O W L E D G M E N T S

    This work was supported by a Royal Childrens HospitalFoundation Grant (922-034), which was sponsored by theWoolworths Care for Kids campaign. The author wishes tothank Dr Katherine Arden for critical review of this manu-script.

    R E F E R E N C E S

    1. Freymuth F, Eugene G, Vabret A et al. Detection of res-piratory syncytial virus by reverse transcription-PCR andhybridization with a DNA enzyme immunoassay. J Clin

    Microbiol 1995; 33: 33523355.2. Mullis KB, Faloona F. Specific synthesis of DNA in vitro

    via a polymerase-catalysed chain reaction. Meth Enzymol1987; 155: 335350.

    3. Niubo J, Perez JL, Carvajal A, Ardanuy C, Martin R.Effect of delayed processing of blood samples on per-formance of cytomegalovirus antigenemia assay. J Clin

    Microbiol 1994; 32: 11191120.4. McAdam AJ. Discrepant analysis: how can we test a test?

    J Clin Microbiol 2000; 38: 20272029.5. Sternberg M. Discrepant analysis is still at large. J Clin

    Microbiol 2001; 39: 826827.6. Jeffery KJM, Read SJ, Peto TEA, Mayon-White RT,

    Bangham CRM. Diagnosis of viral infections of the cen-tral nervous system: clinical interpretation of PCR results.Lancet 1997; 349: 313317.

    7. Pfaffl MW, Horgan GW, Dempfle L. Relative expressionsoftware tool (REST) for group-wise comparison andstatistical analysis of relative expression results in real-time PCR. Nucleic Acids Res 2002; 30: e36.

    8. Bustin SA. Absolute quantification of mRNA using real-time reverse transcription polymerase chain reactionassays. J Mol Endocrinol 2000; 25: 169193.

    9. Nagano M, Kelly PA. Tissue distribution and regula-tion of rat prolactin receptor gene expression. J Biol Chem1994; 269: 1333713345.

    10. Gause WC, Adamovicz J. The use of the PCR to quanti-tate gene expression. PCR Meth Appl 1994; 3: S123135.

    11. Whelan AC, Persing DH. The role of nucleic acid

    amplification and detection in the clinical microbiologylaboratory. Annu Rev Genet 1996; 50: 349373.12. Carman WF, Wallace LA, Walker J et al. Rapid virological

    surveillance of community influenza infection in generalpractice. BMJ 2000; 321: 736737.

    13. Ogilvie M. Molecular techniques should not now replacecell culture in diagnostic virology laboratories. Rev Med2001; 11: 351554.

    14. Biel SS, Madeley D. Diagnostic virologythe need forelectron microscopy: a discussion paper. J Clin Virol 2001;22: 19.

    15. Sintchenko V, Iredell JR, Gilbert GL. Is it time to replacethe petri dish with PCR? Application of culture-inde-pendent nucleic acid amplification in diagnostic bac-teriology: expectations and reality. Pathology 1999; 31:

    436439.16. Ellis JS, Zambon MC. Molecular diagnosis of influenza.

    Rev Med 2002; 12: 375389.17. Clarke SC, Reid J, Thom L, Denham BC, Edwards GFS.

    Laboratory confirmation of meningococcal disease inScotland, 19939. J Clin Pathol 2002; 55: 3236.

    18. Johnson JR. Development of polymerase chain reaction-based assays for bacterial gene detection. J Microbiol Meth2000; 41: 201209.

    19. Kidd IM, Clark DA, Emery VC. A non-radioisotopicquantitative competitive polymerase chain reactionmethod: application in measurement of human herpes-virus 7 load. J Virol Meth 2000; 87: 177181.

    20. Holland PM, Abramson RD, Watson R, Gelfand DH.Detection of specific polymerase chain reaction product

    by utilizing the 5)

    3 exonuclease activity of Thermusaquaticus. Proc Natl Acad Sci USA 1991; 88: 72767280.

    21. van der Vliet GME, Hermans CJ, Klatser PR. Simplecolorimetric microtiter plate hybridization assay fordetection of amplified Mycobacterium leprae DNA. J Clin

    Microbiol 1993; 31: 665670.22. Mackay IM, Metharom P, Sloots TP, Wei MQ. Quanti-

    tative PCR-ELAHA for the determination of retroviralvector transduction efficiency. Mol Ther 2001; 3: 801807.

    23. Guatelli JC, Gingeras TR, Richman DD. Nucleic acidamplification in vitro: detection of sequences with lowcopy numbers and application to diagnosis of humanimmunodeficiency virus type 1 infection. Clin MicrobiolRev 1989; 2: 217226.

    24. Lomeli H, Tyagi S, Pritchard CG, Lizardi PM, KramerFR. Quantitative assays based on the use of repli-catable hybridization probes. Clin Chem 1989; 35: 18261831.

    25. Cockerill FR III, Smith TF. Rapid-cycle real-time PCR: arevolution for clinical microbiology. ASM News 2002; 68:7783.

    26. Lee LG, Connell CR, Bloch W. Allelic discrimination bynick-translation PCR with fluorogenic probes. Nucleic

    Acids Res 1993; 21: 37613766.27. Livak KJ, Flood SJA, Marmaro J, Giusti W, Deetz K.

    Oligonucleotides with fluorescent dyes at opposite ends

    Mackay Real-time PCR in the microbiology laboratory 203

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    15/23

    provide a quenched probe system useful for detectingPCR product and nucleic acid hybridization. PCR Meth

    Appl 1995; 4: 357362.28. Heid CA, Stevens J, Livak KJ, Williams PM. Real time

    quantitative PCR. Genet Meth 1996; 6: 986994.29. Gibson UEM, Heid CA, Williams PM. A novel method

    for real time quantitative RT-PCR. Genome Res 1996; 6:9951001.30. Niesters HG, van Esser J, Fries E, Wolthers KC, Cornel-

    issen J, Osterhaus AD. Development of a real-timequantitative assay for detection of EpsteinBarr virus.

    J Clin Microbiol 2000; 38: 712715.31. Read SJ. Recovery efficiencies of nucleic acid extraction

    kits as measured by quantitative LightCyclerTM PCR.J Clin Pathol 2001; 54: 8690.

    32. Biel SS, Held TK, Landt O et al. Rapid quantification anddifferentiation of human polyomavirus DNA in undi-luted urine from patients after bone marrow transplan-tation. J Clin Microbiol 2000; 38: 36893695.

    33. Petrik J, Pearson GJM, Allain J-P. High throughput PCRdetection of HCV based on semiautomated multisample

    RNA capture. J Virol Meth 1997; 64: 147159.34. Klein D. Quantification using real-time PCR technology:

    applications and limitations. Trend Mol Med 2002; 8: 257260.

    35. Ahmed FE. Detection of genetically modified organismsin foods. Trends Biotechnol 2002; 20: 215223.

    36. Mhlanga MM, Malmberg L. Using molecular beacons todetect single-nucleotide polymorphisms with real-timePCR. Methods 2001; 25: 463471.

    37. Matthews JA, Kricka LJ. Analytical strategies for the useof DNA probes. Anal Biochem 1988; 169: 125.

    38. Wittwer CT, Fillmore GC, Garling DJ. Minimizing thetime required for DNA amplification by efficient heattransfer to small samples. Anal Biochem 1990; 186: 328331.

    39. Wittwer CT, Ririe KM, Andrew RV, David DA, Gundry

    RA, Balis UJ. The LightCyclerTM

    : a microvolume multi-sample fluorimeter with rapid temperature control. Bio-techniques 1997; 22: 176181.

    40. Nitsche A, Steuer N, Schmidt CA et al. Detection ofhuman cytomegalovirus DNA by real-time quantitativePCR. J Clin Microbiol 2000; 38: 27342737.

    41. Lunge VR, Miller BJ, Livak KJ, Batt CA. Factors affectingthe performance of 5 nuclease PCR assays for Listeriamonocytogenes detection.J Microbiol Meth 2002; 51: 361368.

    42. Gyllensten UB, Erlich HA. Generation of single-strandedDNA by the polymerase chain reaction and its applica-tion to direct sequencing of the HLA-DQA locus. ProcNatl Acad Sci USA 1988; 85: 76527656.

    43. Didenko VV. DNA probes using fluorescence resonanceenergy transfer (FRET): designs and applications. Bio-

    techniques 2001; 31: 11061121.44. Stryer L, Haugland RP. Energy transfer: a spectroscopic

    ruler. Proc Natl Acad Sci USA 1967; 58: 719726.45. Heller MJ, Morrison LE. In: Kingsbury DT, Falkow S, eds.

    Rapid detection and identification of infectious agents. NewYork: Academic Press, 1985: 245256.

    46. Clegg RM. Fluorescence resonance energy transfer andnucleic acids. Meth Enzymol 1992; 211: 353388.

    47. Forster T. Zwischenmolekulare energiewanderung undfluoreszenz. Ann Phys 1948; 6: 5575.

    48. Selvin P. Fluorescence resonance energy transfer. MethEnzymol 1995; 246: 300334.

    49. Higuchi R, Fockler C, Dollinger G, Watson R. Kinetic PCRanalysis: real-time monitoring of DNA amplificationreactions. Biotechnology (NY) 1993; 11: 10261030.

    50. Martell M, Gomez J, Esteban JI et al. High-through-put real-time reverse transcription-PCR quantitationof hepatitis C virus RNA. J Clin Microbiol 1999; 37: 327

    332.51. Whitcombe D, Theaker J, Guy SP, Brown T, Little S.Detection of PCR products using self-probing ampliconsand fluorescence. Nat Biotechnol 1999; 17: 804807.

    52. Mackay IM, Arden KE, Nitsche A. Real-time PCR invirology. Nucleic Acids Res 2002; 30: 12921305.

    53. Egholm M, Buchardt O, Christensen L et al. PNAhybridizes to complementary oligonucleotides obeyingthe WatsonCrick hydrogen bonding rules. Nature 1993;365: 566568.

    54. Cardullo RA, Agrawai S, Flores C, Zamecnik PC, WolfDE. Detection of nucleic acid hybridization by nonradi-ative fluorescence resonance energy transfer. Proc Natl

    Acad Sci USA 1988; 85: 87908794.55. Wittwer CT, Herrmann MG, Moss AA, Rasmussen RP.

    Continuous fluorescence monitoring of rapid cycle DNAamplification. Biotechniques 1997; 22: 130138.

    56. Li Q, Luan G, Guo Q, Liang J. A new class of homogen-eous nucleic acid probes based on specific displacementhybridization. Nucleic Acids Res 2002; 30: e5.

    57. Stender H, Fiandaca M, Hyldig-Nielsen JJ, Coull J. PNAfor rapid microbiology. J Microbiol Meth 2002; 48: 117.

    58. Fiandaca MJ, Hyldig-Nielsen JJ, Gildea BD, Coull JM.Self-reporting PNADNA primers for PCR analysis.Genome Res 2001; 11: 609611.

    59. Svanvik N, Westman G, Wang D, Kubista M. Light-upprobes. Thiazole orange-conjugated peptide nucleic acidfor the detection of target nucleic acid in homogeneoussolution. Anal Biochem 2001; 281: 2635.

    60. Isacsson J, Cao H, Ohlsson L et al. Rapid and specific

    detection of PCR products using light-up probes. Mol CellProbes 2000; 14: 321328.

    61. Svanvik N, Sehlstedt U, Sjoback R, Kubista M. Detectionof PCR products in real time using light-up probes. AnalBiochem 2000; 287: 179182.

    62. French DJ, Archard CL, Brown T, McDowell DG. HyBea-conTM probes: a newtool for DNAsequence detection andallele discrimination. Mol Cell Probes 2001; 15: 363374.

    63. French DJ, Archard CL, Andersen MT, McDowell DG.Ultra-rapid analysis using HyBeaconTM probes and directPCR amplification from saliva. Mol Cell Probes 2002; 16:319326.

    64. Afonina IA, Reed MW, Lusby E, Shishkina IG, BelousovYS. Minor groove binder-conjugated DNA probes forquantitative DNA detection by hybridization-triggered

    fluorescence. Biotechniques 2002; 32: 940949.65. Kutyavin IV, Afonina IA, Mills A et al. 3-Minor groove

    binder-DNA probes increase sequence specificity at PCRextension temperatures. Nucleic Acids Res 2000; 28: 655661.

    66. Afonina IA, Sanders S, Walburger D, Belousov YS.Accurate SNP typing by real-time PCR. Pharmagenomics2002; JanFeb: 4854.

    67. Todd AV, Fuery CJ, Impey HL, Applegate TL, HaughtonMA. DzyNA-PCR. Use of DNAzymes to detect andquantify nucleic acid sequences in a real-time fluorescentformat. Clin Chem 2000; 46: 625630.

    204 Clinical Microbiology and Infection, Volume 10 Number 3, March 2004

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    16/23

    68. Tyagi S, Bratu DP, Kramer FR. Multicolor molecularbeacons for allele discrimination. Nat Biotechnol 1998; 16:4953.

    69. Tyagi S, Kramer FR. Molecular beacons: probes thatfluoresce upon hybridization. Nat Biotechnol 1996; 14:303308.

    70. Nutiu R, Li Y. Tripartite molecular beacons. Nucleic AcidsRes 2002; 30: e94.71. Nazarenko I, Lowe B, Darfler M, Ikonomi P, Schuster D,

    Raschtchian A. Multiplex quantitative PCR using self-quenched primers labeled with a single fluorophore.Nucleic Acids Res 2002; 30: e37.

    72. Celi FS, Mentuccia D, Proietti-Pannunzi L, di Gioia CRT,Andreoli M. Preparing poly-A-containing RNA internalstandards for multiplex competitive RT-PCR. Biotech-niques 2000; 29: 454458.

    73. Alexandre I, Zammatteo N, Ernest I et al. Quantitativedetermination of CMV DNA using a combination ofcompetitive PCR amplification and sandwich hybridiza-tion. Biotechniques 1998; 25: 676683.

    74. Zimmermann K, Manhalter JW. Technical aspects of

    quantitative competitive PCR. Biotechniques 1996; 21: 268279.

    75. Brightwell G, Pearce M, Leslie D. Development of inter-nal controls for PCR detection of Bacillus anthracis. MolCell Probes 1998; 12: 367377.

    76. Glasel JA. Validity of nucleic acid purities monitored by260nm280nm absorbance ratios. Biotechniques 1995; 18:6263.

    77. Bagnarelli P, Menzo S, Valenza A et al. Quantitativemolecular monitoring of human immunodeficiency virustype 1 activity during therapy with specific antiretroviralcompounds. J Clin Microbiol 1995; 33: 1623.

    78. Wang Z, Spadoro J. Determination of target copy numberof quantitative standards used in PCR based diagnosticassays. In: Ferre F, ed. Gene quantification. Boston: Bir-

    khauser, 1998: 3134.79. Rodrigo AG, Goracke PC, Rowhanian K, Mullins JI.

    Quantitation of target molecules from polymerase chainreaction-based limiting dilution assays. AIDS Res HumRetrovirus 1997; 13: 737742.

    80. Taswell C. Limiting dilution assays for the determinationof immunocompetent cell frequencies. J Immunol 1981;126: 16141619.

    81. Sykes PJ, Brisco MJ, Snell LE et al. Minimal residual dis-ease in childhood acute lymphoblastic leukaemia quan-tified by aspirate and trephine: is the disease multifocal?Br J Haematol 1998; 103: 6065.

    82. Niesters HGM. Quantitation of viral load using real-time amplification techniques. Methods 2001; 25: 419429.

    83. Borson ND, Strausbauch MA, Wettstein PJ, Oda RP,Johnston SL, Landers JP. Direct quantitation ofRNA transcripts by competitive single-tube RT-PCRand capillary electrophoresis. Biotechniques 1998; 25:130137.

    84. Gelmini S, Orlando C, Sestini R et al. Quantitativepolymerase chain reaction-based homogeneous assaywith fluorogenic probes to measure c-cerB-2 oncogeneamplification. Clin Chem 1997; 43: 752758.

    85. Chehab FF, Doherty M, Cai S, Kan YW, Cooper S, RubinEM. Detection of sickle cell anaemia and thalassaemias.Nature 1987; 329: 293294.

    86. Reiss RA, Rutz B. Quality control PCR: a method fordetecting inhibitors of Taq DNA polymerase. Biotech-niques 1999; 27: 920926.

    87. Orlando C, Pinzani P, Pazzagli M. Developments inquantitative PCR. Clin Chem Lab Med 1998; 36: 255269.

    88. Moller A, Jansson JK. Quantification of genetically tagged

    cyanobacteria in Baltic sea sediment by competitive PCR.Biotechniques 1997; 22: 512518.89. Hall LL, Bicknell GR, Primrose L, Pringle JH, Shaw JA,

    Furness PN. Reproducibility in the quantification ofmRNA levels by RT-PCR-ELISA and RT competitive-PCR-ELISA. Biotechniques 1998; 24: 652657.

    90. Aberham C, Pendl C, Gross P, Zerlauth G, Gessner M. Aquantitative, internally controlled real-time PCR assay forthe detection of parvovirus B19 DNA. J Virol Meth 2001;92: 183191.

    91. Kearns AM, Guiver M, James V, King J. Developmentand evaluation of a real-time quantitative PCR for thedetection of human cytomegalovirus. J Virol Meth 2001;95: 121131.

    92. Gruber F, Falkner FG, Dorner F, Hammerle T. Quantita-

    tion of viral DNA by real-time PCR applying duplexamplification, internal standardization, and two-colourfluorescence detection. Appl Environ Microbiol 2001; 67:28372839.

    93. Selvey S, Thompson EW, Matthaei K, Lea RA, Irving MG,Griffiths LR. b-Actinan unsuitable internal control forRT-PCR. Mol Cell Probes 2001; 15: 307311.

    94. Thellin O, Zorzi W, Lakaye B et al. Housekeeping genesas internal standards: use and limits. J Biotechnol 1999; 75:291295.

    95. Freeman WM, Walker SJ, Vrana KE. QuantitativeRT-PCR: pitfalls and potential. Biotechniques 1999; 26:112125.

    96. Becker-Andre M, Hahlbrock K. Absolute mRNA quanti-fication using the polymerase chain reaction (PCR). A

    novel approach by a PCR aided transcript titration assay(PATTY). Nucleic Acids Res 1989; 17: 94379447.

    97. Clementi M, Menzo S, Manzin A, Bagnarelli P. Quanti-tative molecular methods in virology. Arch Virol 1995;140: 15231539.

    98. Gilliland G, Perrin S, Bunn HF. PCR protocols: a guide tomethods and applications. San Diego: Academic Press, 1990:6069.

    99. Siebert PD, Larrick JW. Competitive PCR. Nature 1992;359: 557558.

    100. Locatelli G, Santoro F, Veglia F, Gobbi A, Lusso P,Malnati MS. Real-time quantitative PCR for humanherpesvirus 6 DNA. J Clin Microbiol 2000; 38: 40424048.

    101. Wall SJ, Edwards DR. Quantitative reverse transcription-polymerase chain reaction (RT-PCR): a comparison of

    primer-dropping, competitive, and real-time RT-PCRs.Anal Biochem 2002; 300: 269273.

    102. Tanaka N, Kimura H, Iida K et al. Quantitative analysis ofcytomegalovirus load using a real-time PCR assay. J MedVirol 2000; 60: 455462.

    103. Liu W, Saint DA. A new quantitative method of real timereverse transcription polymerase chain reaction assaybased on simulation of polymerase chain reaction kinet-ics. Anal Biochem 2002; 302: 5259.

    104. Wilhelm J, Hahn M, Pingoud A. Influence of DNA targetmelting behavior on real-time PCR quantification. ClinChem 2001; 46: 17381743.

    Mackay Real-time PCR in the microbiology laboratory 205

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    17/23

    105. Jung R, Soondrum K, Neumaier M. Quantitative PCR.Clin Chem Lab Med 2000; 38: 833836.

    106. Ishiguro T, Saitoh J, Yawata H, Yamagishi H, Iwasaki S,Mitoma Y. Homogeneous quantitative assay of hepatitisC virus RNA by polymerase chain reaction in the pres-ence of a fluorescent intercalater. Anal Biochem 1995; 229:

    207213.107. Kimura H, Morita M, Yabuta Y et al. Quantitative analysisof EpsteinBarr virus load by using a real-time PCRassay. J Clin Microbiol 1999; 37: 132136.

    108. Najioullah F, Thouvenot D, Lina B. Development of areal-time PCR procedure including an internal control forthe measurement of HCMV viral load. J Virol Meth 2001;92: 5564.

    109. Ryncarz AJ, Goddard J, Wald A, Huang M-L, Roizman B,Corey L. Development of a high-throughput quantitativeassay for detecting herpes simplex virus DNA in clinicalsamples. J Clin Microbiol 1999; 37: 19411947.

    110. Monopoeho S, Mignotte B, Schwartzbrod L et al. Quan-tification of enterovirus RNA in sludge samples usingsingle tube real-time RT-PCR. Biotechniques 2000; 29:

    8893.111. Alexandersen S, Oleksiewicz MB, Donaldson AI. The

    early pathogenesis of foot-and-mouth disease in pigsinfected by contact: a quantitative time-course studyusing TaqMan RT-PCR. J Gen Virol 2001; 82: 747755.

    112. Abe A, Inoue K, Tanaka T et al. Quantitation of hepatitis Bvirus genomic DNA by real-time detection PCR. J Clin

    Microbiol 1999; 37: 28992903.113. Brechtbuehl K, Whalley SA, Dusheiko GM, Saunders

    NA. A rapid real-time quantitative polymerase chainreaction for hepatitis B virus. J Virol Meth 2001; 93:105113.

    114. Moody A, Sellers S, Bumstead N. Measuring infectiousbursal disease virus RNA in blood by multiplex real-timequantitative RT-PCR. J Virol Meth 2000; 85: 5564.

    115. Weinberger KM, Wiedenmann E, Bohm S, Jilg W. Sensi-tive and accurate detection of hepatitis B virus DNAusing a kinetic fluorescence detection system (TaqManPCR). J Virol Meth 2000; 85: 7582.

    116. Schaade L, Kockelkorn P, Ritter K, Kleines M. Detectionof cytomegalovirus DNA in human specimens by Light-Cycler PCR. J Clin Microbiol 2000; 38: 40064009.

    117. Kawai S, Yokosuka O, Kanda T, Imazeki F, Maru Y,Saisho H. Quantification of hepatitis C virus by TaqManPCR: comparison with HCV amplicor monitor assay. J

    Med Virol 1999; 58: 121126.118. Schutten M, van den Hoogen B, van der Ende ME,

    Gruters RA, Osterhaus ADME, Niesters HGM. Devel-opment of a real-time quantitative RT-PCR for thedetection of HIV-2 RNA in plasma. J Virol Meth 2000; 88:

    8187.119. Kearns AM, Turner AJL, Taylor CE, George PW, Freeman

    R, Gennery AR. LightCycler-based quantitative PCR forrapid detection of human herpesvirus 6 DNA in clinicalmaterial. J Clin Microbiol 2001; 39: 30203021.

    120. Kupferschmidt O, Kruger D, Held TK, Ellerbrok H,Siegert W, Janitschke K. Quantitative detection of Toxo-plasma gondii DNA in human body fluids by TaqManpolymerase chain reaction. Clin Microbiol Infect 2001; 7:120124.

    121. Kennedy MM, Lucas SB, Russell-Jones R et al. HHV8and female Kaposis sarcoma. J Pathol 1997; 183: 447452.

    122. Capone RB, Pai SI, Koch WM, Gillison ML. Detection andquantitation of human papillomavirus (HPV) DNA in thesera of patients with HPV-associated head and necksquamous cell carcinoma. Clin Cancer Res 2001; 6: 41714175.

    123. Leutenegger CM, Klein D, Hofmann-Lehmann R et al.

    Rapid feline immunodeficiency virus provirus quantita-tion by polymerase chain reaction using the TaqManfluorogenic real-time detection system. J Virol Meth 1999;78: 105116.

    124. Smith IL, Halpin K, Warrilow D, Smith GA. Developmentof a fluorogenic RT-PCR assay (TaqMan) for the detectionof Hendra virus. J Virol Meth 2001; 98: 3340.

    125. van Elden LJR, Nijhuis M, Schipper P, Schuurman R, vanLoon AM. Simultaneous detection of influenza viruses Aand B using real-time quantitative PCR. J Clin Microbiol2001; 39: 196200.

    126. Lanciotti RS, Kerst AJ, Nasci RS et al. Rapid detectionof west nile virus from human clinical specimens, field-collected mosquitoes, and avian samples by a TaqManreverse transcriptase-PCR assay. J Clin Microbiol 2000; 38:

    40664071.127. Bernard PS, Lay MJ, Wittwer CT. Integrated amplification

    and detection of the C677T point mutation in the meth-yltetrahydrofolate reductase gene by fluorescence reson-ance energy transfer and probe melting curves. AnalBiochem 1998; 255: 101107.

    128. Chamberlain JS, Gibbs RA, Ranier JE, Nguyen PN, Cas-key CT. Deletion screening of the Duchenne musculardystrophy locus via multiplex DNA amplification.Nucleic Acids Res 1988; 16: 1114111156.

    129. Elnifro EM, Ashshi AM, Cooper RJ, Klapper PE. Multi-plex PCR: optimization and application in diagnosticvirology. Clin Microbiol Rev 2000; 13: 559570.

    130. Tyagi S, Marras SAE, Kramer FR. Wavelength-shiftingmolecular beacons. Nat Biotechnol 2000; 18: 11911196.

    131. Nazarenko I, Pires R, Lowe B, Obaidy M, Raschtchian A.Effect of primary and secondary structure of oligode-oxyribonucleotides on the fluorescent properties of con-jugated dyes. Nucleic Acids Res 2002; 30: 20892195.

    132. Espy MJ, Uhl JR, Mitchell PS et al. Diagnosis of herpessimplex virus infections in the clinical laboratory byLightCycler PCR. J Clin Microbiol 2000; 38: 795799.

    133. Nicolas L, Milon G, Prina E. Rapid differentiation of oldworld Leishmania species by LightCycler polymerasechain reaction and melting curve analysis. J Microbiol

    Meth 2002; 51: 295299.134. Wittwer CT, Herrmann MG, Gundry CN, Elenitoba-

    Johnson KSJ. Real-time multiplex PCR assays. Methods2001; 25: 430442.

    135. Espy MJ, Ross TK, Teo R et al. Evaluation of LightCycler

    PCR for implementation of laboratory diagnosis of her-pes simplex virus infections. J Clin Microbiol 2000; 38:31163118.

    136. Schalasta G, Arents A, Schmid M, Braun RW, Enders G.Fast and type-specific analysis of herpes simplex virustypes 1 and 2 by rapid PCR and fluorescence melting-curve-analysis. Infection 2000; 28: 8591.

    137. Loparev VN, McCaustland K, Holloway BP, Krause PR,Takayama M, Schmid DS. Rapid genotyping of varicella-zoster virus vaccine and wild-type strains with fluoro-phore-labeled hybridization probes. J Clin Microbiol 2000;38: 43154319.

    206 Clinical Microbiology and Infection, Volume 10 Number 3, March 2004

    2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10, 190212

  • 8/2/2019 Real-Time PCR in the Microbiology Laboratory

    18/23

    138. Read SJ, Mitchell JL, Fink CG. LightCycler multiplex PCRfor the laboratory diagnosis of common viral infections ofthe central nervous system. J Clin Microbiol 2001; 39:30563059.

    139. Whiley DM, Mackay IM, Sloots TP. Detection anddifferentiation of human polyomaviruses JC and BK

    by LightCycler PCR. J Clin Microbiol 2001; 39: 43574361.140. Schutz E, von Ashen N, Oellerich M. Genotyping of eightthiopurine methyltransferase mutations: three-colormultiplexing, two-colorshared anchor, and fluores-cence-quenching hybridization probe assays based onthermodynamic nearest-neighbour probe design. ClinChem 2000; 46: 17281737.

    141. Bohling SD, King TC, Wittwer CT, Elenitoba-JohnsonKSJ. Rapid simultaneous amplification and detection ofthe MBRJH chromosomal translocation by fluorescencemelting curve analysis. Am J Pathol 1999; 154: 97103.

    142. Lay MJ, Wittwer CT. Real-time fluorescence genotypingof factor V Leiden during rapid-cycle PCR. Clin Chem1997; 43: 22622267.

    143. Herrmann MG, Dobrowolski SF, Wittwer CT. Rapid

    b-globin genotyping by multiplexing probe meltingtemperature and color. Clin Chem 2000; 46: 425429.

    144. Lee LG, Livak KJ, Mullah B, Graham RJ, Vinayak RS,Woudenberg TM. Seven-color, homogeneous detection ofsix PCR products. Biotechniques 1999; 27: 342349.

    145. Gundry CN, Bernard PS, Herrmann MG, Reed GH, Wit-twer CT. Rapid F508del and F508C assay using fluorescenthybridization probes. Genet Test 1999; 3: 365370.

    146. Bernard PS, Wittwer CT. Homogeneous amplificationand variant detection by fluorescent hybridizationprobes. Clin Chem 2000; 46: 147148.

    147. Elenitoba KSJ, Bohling SD, Wittwer CT, King TC. Multi-plex PCR by multicolor fluorimetry and fluorescencemelting curve analysis. Nat Med 2001; 7: 249253.

    148. Vet JAM, Majithia AR, Marras SAE et al. Multiplex

    detection of four pathogenic retroviruses using molecularbeacons. Proc Natl Acad Sci USA 1999; 96: 63946399.

    149. Quereda C, Corral I, Laguna Fet al. Diagnostic utility of amultiplex herpesvirus PCR assay performed with cere-brospinal fluid from human immunodeficiency virus-infected patients with neurological disorders. J Clin

    Microbiol 2000; 38: 30613067.150. Weigl JAI, Puppe W, Grondahl B, Schmitt H-J. Epide-

    miological investigation of nine respiratory pathogens inhospitalized children in Germany using multiplexreverse-transcriptase polymerase chain reaction. Eur JClin Microbiol Infect Dis 2000; 19: 336343.

    151. Stockton J, Ellis JS, Saville M, Clewley JP, Zambon MC.Multiplex PCR for typing and subtyping influenza andrespiratory syncytial viruses. J Clin Microbiol 1998; 36:

    29902995.152. Kehl SC, Henrickson KJ, Hua W, Fan J. Evaluation of the

    hexaplex assay for detection of respiratory viruses inchildren. J Clin Microbiol 2001; 39: 16961701.

    153. Echevarria JE, Erdman DD, Swierkosz EM, Holloway BP,Anderson LJ. Simultaneous detection and identificationof human parainfluenza viruses 1, 2, and 3 from clinicalsamples by multiplex PCR. J Clin Microbiol 1998; 36: 13881391.

    154. Henegariu O, Heerema NA, Dloughy SR, Vance GH,Vogt PH. Multiplex PCR: critical parameters and step-by-step protocol. Biotechniques 1997; 23: 504511.

    155. Schalasta G, Eggers M, Schmid M, Enders G. Analysis ofhuman cytomegalovirus DNA in urines of newborns andinfants by means of a new ultrarapid real-time PCR-system. J Clin Virol 2000; 19: 175185.

    156. Kato T, Mizokami M, Mukaide M et al. Development of aTT virus DNA quantification system using real-time

    detection PCR. J Clin Microbiol 2000; 38: 9498.157. Zerr DM, Huang M-L, Corey L, Erickson M, Parker HL,Frenkel LM. Sensitive method for detection of humanherpesvirus 6 and 7 in saliva collected in field studies. JClin Microbiol 2000; 38: 19811983.

    158. Furuta Y, Ohtani F, Sawa H, Fukuda S, Inuyama Y.Quantitation of varicella-zoster virus DNA in patientswith Ramsay Hunt syndrome and zoster sine herpete. JClin Microbiol 2001; 39: 28562859.

    159. Jordens JZ, Lanham S, Pickett MA, Amarasekara S,Aberywickrema I, Watt PJ. Amplification with molecularbeacon primers and reverse line blotting for the detectionand typing of human papillomaviruses. J Virol Meth 2000;89: 2937.

    160. Aldous EW, Collins MS, McGoldrick A, Alexander DJ.

    Rapid pathotyping of Newcastle disease virus (NDV)using fluorogenic probes in a PCR assay. Vet Microbiol2001; 80: 201212.

    161. Kennedy MM, Lucas SB, Jones RR et al. HHV8 andKaposis sarcoma: a time cohort study. J Clin Pathol 1997;50: 96100.

    162. Kennedy MM, Cooper K, Howells DD et al. Identificationof HHV8 in early Kaposis sarcoma: implications forKaposis sarcoma pathogenesis. J Clin Pathol 1998; 51:1420.

    163. OLeary JJ, Kennedy M, Luttich K et al. Localisation ofHHV-8 in AIDS related lymphadenopathy. J Clin Pathol2000; 53: 4347.

    164. OLeary J, Kennedy M, Howells D et al. Cellularlocalisation of HHV-8 in Castlemans disease: is there a

    link with lymphnode vascularity? J Clin Pathol 2000; 53:6976.

    165. Lo YMD, Chan LYS, Lo K-W et al. Quantitative analysisof cell-free EpsteinBarr virus DNA in plasma of patientswith nasopharyngeal carcinoma. Cancer Res 1999; 59:11881191.

    166. Josefsson A, Livak K, Gyllensten U. Detection andquantitation of human papillomavirus by using thefluorescent 5 exonuclease assay. J Clin Microbiol 1999; 37:490496.

    167. Swan DC, Tucker RA, Holloway BP, Icenogle JP. A sen-sitive, type-specific, fluorogenic probe assay for detectionof human papillomavirus DNA. J Clin Microbiol 1997; 35:886891.

    168. Lanham S, Herbert A, Watt P. HPV detection and

    measurement of HPV-16, telomerase, and survivingtranscripts in colposcopy clinic patients. J Clin Pathol2001; 54: 304308.

    169. MacKenzie J, Gallagher A, Clayton RA et al. Screening forherpesvirus genomes in common acute lymphoblasticleukemia. Leukemia 2001; 15: 415421.

    170. Jabs WJ, Hennig H, Kittel M et al. Normalized quantifi-cation by real-time PCR of EpsteinBarr virus load inpatients at risk for posttransplant lymphoproliferativedisorders. J Clin Microbiol 2001