Anda di halaman 1dari 6

Clinical CMN

Microbiology
Stay Current...
Stay Informed.

N e w s l e t t e r

CMN Digital PCR in the Clinical Microbiology Laboratory:


Vol. 40, No. 4
February 15, 2018
Another Tool on the Molecular Horizon
www.cmnewsletter.com Eleanor A. Powell, Ph.D.1 and N. Esther Babady, Ph.D., D(ABMM),1,2 1Clinical Microbiology Service,
Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York,
2
Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York,
I n Th is Issu e
New York
27 Digital PCR in the Clinical
Microbiology Laboratory: Abstract
Another Tool on the Digital PCR was first described in 1999. Improvements in instrumentation and reagents have increased
Molecular Horizon its routine use in molecular pathology laboratories. For clinical microbiology laboratories, digital
PCR applications could cover a broad range of purposes, from absolute pathogen quantification to
32 A Farewell Tribute to
Retiring CMN Editors and
the detection of rare mutations that confer resistance to antibiotics. In this review, we describe several
their Contribution to potential applications of digital PCR in the clinical microbiology laboratory. As this method contin-
Clinical Microbiology ues to evolve, digital PCR may become an important tool on the horizon for clinical and molecular
microbiology laboratories.

Introduction was first coined in a 1999 report by Vogelstein


and Kinzler at Johns Hopkins [3, 4]. Building on
The introduction of conventional endpoint poly-
previous works that described limiting-dilution
merase chain reaction (PCR) and real-time PCR
and single-molecule amplification, Vogelstein
in clinical microbiology laboratories was transfor-
and Kinzler developed a method to amplify an
mative, allowing the identification of pathogens
individual template DNA molecule in a separate
within a few hours when such tasks traditionally
reaction and, with the use of fluorescent probes,
took several days or weeks to complete. The
detect and count the amplified reactions, yield-
field of nucleic acid amplification technology
ing a “digital” signal (Fig. 1). They applied this
is continually evolving to improve the sensitiv-
method to detect low concentrations of a rare
ity of nucleic acid detection, reduce the risk of
Ras oncogene mutation in a stool sample from a
cross-contamination, and allow accurate quan-
patient with colorectal cancer. Since these early
tification. Thus, digital PCR (dPCR) was born
reports, the number of publications that describe
out of a need to amplify and accurately quantify
the use of dPCR have increased significantly, with
a single molecule or a rare allele that is present
most applications found in the fields of human
in excess of wild-type alleles. An allele is one of
genetics and oncology. This increase was partly
several alternative forms of the same gene or the
due to improvements in a variety of instrument
same genetic locus.
microfluidics and partitioning chemistries that
Corresponding author: N. Esther Reports of single-molecule amplifications were resulted in simplification of the original proto-
Babady, Ph.D., 327 East 64th published as early as the late 1980s, a few years col developed by Vogelstein and Kinzler. Most
Street CLM 522, New York, NY after the description of PCR. Single-molecule recently, dPCR has provided value to an increas-
10065. Tel.: 212-639-8179. amplification was made possible by physically ing number of applications in clinical microbi-
E-mail: babadyn@mskcc.org separating single cells or by performing limit- ology (Table 1). This review summarizes and
0196-4399/©2018 Elsevier Inc. ing dilution of samples prior to amplification discusses the applications of dPCR in the clinical
All rights reserved and detection [1,2]. The term “digital” PCR microbiology laboratory.

Clinical Microbiology Newsletter 40:4,2018 | ©2018 Elsevier 27


Figure 1. dPCR workflow.
Sample (total nucleic acids
[NA]) is divided into multiple
partitions containing either 0
or 1 template NA. Amplification
of a single-molecule template
occurs within each partition.
If the assay is designed to
detect more than one target, a
different fluorescent probe (e.g.,
FAM or VIC) can be used for the
detection of amplified template.
The data are presented in
either a one-dimensional (1D)
scatterplot with all positive
partitions above an established
threshold or a 2D scatterplot
where the number of partitions
positive for one-target (a and
d) or two targets (c) or empty
partitions (b) can be visualized.

dPCR Technology and Platforms manufacturers have instruments on the market (Table 2). Manu-
facturers of dPCR instruments vary primarily in their approaches
dPCR technology builds on methods that are well established for
to partitioning of the sample. The two main approaches are drop-
conventional and real-time PCR, including nucleic acid extraction,
let and chip partitioning. Of the systems commercially available
amplification of nucleic acids using specific primers, and detection
in the United States, the RainDrop (RainDance Technologies,
of amplified nucleic acids using fluorescent probes (e.g., TaqMan
recently acquired by Bio-Rad Laboratories, Hercules, CA) and
or Molecular Beacons) or DNA intercalating dyes (e.g., SYBR
QX100/200 (Bio-Rad Laboratories) systems use droplet-based
Green or EvaGreen). The workflow for dPCR can be divided into
partitioning, while the Biomark HD (Fluidigm) and QuantStudio
three steps that occur after DNA extraction: (i) sample partition-
3D (ThermoFisher Scientific, Waltham, MA) systems use chip-
ing, (ii) single-molecule amplification, and (iii) signal detection
based partitioning. Other systems available outside the U.S. have
by scatterplot (Fig. 1).
variations on the theme of the chip. For example, the Clarity sys-
Sample partitioning is the process by which a sample (e.g., tem (JNMedsys, Singapore) uses chip-based partitioning, but with
extracted DNA) is diluted and divided into thousands of partitions, a “chip-in-a-tube” design so that amplification can be performed
thereby dividing up the reaction into multiple smaller reactions. on any traditional thermal cycler, while the Naico Geode (Stilla
This step is critical to ensure that each partition ideally contains Technologies, France) uses the Sapphire chip to create crystal-like
either zero or one template molecule. Target amplification is then droplets. Depending on the manufacturer, amplification may be
performed inside each partition simultaneously, resulting in hun- carried out on a specialized thermocycler or on a standard thermo-
dreds to thousands of individual PCRs. Amplified templates are cycler. Detection is carried out on a manufacturer-specific reader.
detected using the fluorescent signal (e.g., 6-carboxyfluorescein
In addition to how partitions are generated, instruments also vary
[FAM] dye or VIC dye), with partitions that contain an amplified
in the number of partitions that are generated, ranging from a
template molecule counted as one (i.e., positive; a single count)
few thousand to several million. The maximum number of par-
and those without amplified template counted as zero (i.e., nega-
titions that can be generated is related to the dynamic range of
tive; no signal is detected). Hence, the total number of positive
the instrument; however, since one of the advantages of dPCR is
partitions reflects the absolute number of targets in the starting
the detection and quantitation of rare targets, a wide linear range
material (Figure 1). Data can be viewed as a single scatterplot, with
may not be necessary.
a threshold for positive set above the background fluorescence
of the negative, or empty, partitions. To account for instances in dPCR versus Real-Time PCR
which more than one template molecule may be present in a par-
dPCR offers several potential benefits over quantitative real-time
tition, a Poisson model mathematical correction is applied during
PCR (qPCR). First, quantification of nucleic acids is absolute,
the analysis. Additionally, using multiplexed dPCR with differ-
negating the need for an external standard curve. This is par-
ent probes targeting different templates, even rare events can be
ticularly useful for pathogens that do not have well-characterized
amplified and detected equally well, since no competition for PCR
reference materials (e.g., World Health Organization [WHO]
reagents with other targets occurs within each partition (Fig. 1).
standards) available. Additionally, with qPCR, differences in
The first commercially available dPCR instrument was released sequence between the materials used for the standard curve and
by Fluidigm (San Francisco, CA) in 2006. Today, several the sample can result in differing primer affinities and different

28 Clinical Microbiology Newsletter 40:4,2018 | ©2018 Elsevier


amplification efficiencies for the standard curve and sample (e.g., Hayden and colleagues further investigated potential causes of dif-
rhinoviruses) [5]. This circumstance can result in artificially high ferences in absolute quantitation by comparing dPCR performance
or low viral loads (for higher affinities and lower affinities, respec- across platforms (Bio-Rad QX200 and RainDance RainDrop)
tively). In contrast, dPCR is less affected by differences in sequence using commercially available CMV analyte-specific reagents, spe-
variation, as partitions with any level of fluorescence are considered cifically, Altona Diagnostics (Hamburg, Germany) RealStar CMV,
positive [5]. Furthermore, compared to qPCR, dPCR is less sus- Abbott Laboratories (Abbott Park, IL) CMV ASR, and Focus
ceptible to inhibitory substances, like those found in feces, heparin, Diagnostics (Cypress, CA) CMV ASR [10]. The authors showed
and SDS, which may allow direct-from-specimen testing or extrac- good correlations across platforms and reagents when measuring
tion methods that are more rapid and/or less labor-intensive [6,7]. CMV standards, though increased variation was observed at low
viral loads. Differences in the lower limit of detection, linearity,
Despite these advantages, compared to qPCR, dPCR has several and overall agreement were both reagent and platform dependent.
drawbacks. dPCR instruments and consumables are generally Hence, interpretation of dPCR data needs to take into consider-
more expensive than those used for qPCR. The workflow for ation multiple variables.
most dPCR platforms has, in some ways, taken a step backward
by requiring multiple steps (partitioning, amplification, and anal- Other studies have compared dPCR to qPCR, and in a recent
ysis of partitions), each with hands-on processing time. In addi- systematic literature review of the application of dPCR to HIV-1
tion, dPCR’s open system is more susceptible to contamination, quantification, dPCR was more accurate, precise, and reproduc-
with risks similar to those with conventional endpoint PCR. The ible than qPCR. Also, though the two methods had equivalent
throughput of dPCR instruments is also generally lower than that sensitivity, the specificity of dPCR was lower due to the presence
of qPCR, particularly for droplet-based technologies. Finally, the of positive droplets in the negative-control partitions [11].
dynamic range of dPCR is usually less than that of qPCR, though
it does vary among instruments. The dynamic range is determined Several studies have reported increased sensitivity of dPCR com-
by the number of partitions, which varies between platforms. The pared to qPCR for a wide range of viruses and sample types,
RainDance platform is capable of 10 million partitions/reactions, including parechovirus in cerebrospinal fluid (CSF), JC virus in
which achieves a 6-log-unit dynamic range [8]. This capability CSF and serum, hepatitis B virus, and H7N9 influenza virus [12-
approaches the 7-log-unit dynamic range that is typical of qPCR. 15]. This increased sensitivity may be due to the partitioning,
Other dPCR platforms have smaller dynamic ranges. which removes the competition between the genetic target and
other extraneous DNA by virtue of the low-copy-number target
Clinical Microbiology Applications in the partition [15]. The increased sensitivity of dPCR may allow
novel applications, like detecting human papillomavirus (HPV)
dPCR has several potential applications for the clinical microbiol- in liquid biopsy fluid. A study of patients with invasive carcinoma
ogy laboratory, where qPCR (aka quantitative real-time PCR) is showed 87% of the patients had detectable HPV using a dPCR
typically used and nucleic acids are quantified by comparing signal assay, while no patients with high-grade cervical intra-epithelial
strength to that of an external standard curve. Although the avail- neoplasia did [16].
ability of the WHO international standards and in vitro diagnostics
(IVD)-cleared qPCR assays has helped decrease the variability in Since dPCR is less susceptible to the effects of inhibitors than is
viral loads observed across laboratories, challenges with compara- qPCR, it can be used to design sensitive assays in more difficult or
bility of viral load testing results still exist, especially when stan- complex matrices. dPCR was used to detect CMV and Epstein-
dards are not available. In light of qPCR variability, dPCR can be Barr virus in formalin-fixed paraffin-embedded tissues from the
used in pathogen quantification, with the ability to provide abso- colonic mucosa of people with HIV [17] and Merkel cell polyoma-
lute quantification instead of quantification relative to a standard virus, with higher sensitivity than qPCR [18]. dPCR was also able
curve. In one study, quantifications of cytomegalovirus (CMV) by to accurately quantify CMV in simple matrices, like Tris buffer and
dPCR and qPCR using standard reference materials across mul- cell lysate, without prior DNA extraction [19]. Avoiding a sepa-
tiple laboratories and platforms were compared; although some rate DNA extraction step would simplify the workflow, remove
differences remained, dPCR demonstrated high reproducibility one possible source of cross-contamination, and remove a source
and decreased inter-laboratory variations compared to qPCR [9]. of inter-assay variability.

Table 1. Applications of dPCR in clinical microbiology


Application Example Reference
Detection Cell-free human papillomavirus DNA in plasma 16
Absolute quantitation Cytomegalovirus quantitation 10
Rare genotype identification Heteroresistance in Mycobacterium tuberculosis 24
Detection of viral genome integration in host genome Chromosomally integrated human herpesvirus 6 20
Reference materials and standards characterization Diversity of viral subpopulations in WHO BK virus standards 33

Clinical Microbiology Newsletter 40:4,2018 | ©2018 Elsevier 29


Current methods to differentiate between cell-free human herpes- dPCR may also be used to provide rapid antimicrobial susceptibil-
virus 6 (HHV-6) and chromosomally integrated HHV-6 include ity results. For these assays, bacteria are exposed to an antibiotic
fluorescent in-situ hybridization and testing of hair follicles for for 15 minutes, and then, the cells are lysed and the bacterial chro-
HHV-6. A group at the University of Washington recently devel- mosomal DNA is partitioned in dPCR, which quantifies bacte-
oped a multiplexed dPCR to measure HHV-6 integration by rial resistance gene replication [26]. While this method has been
quantifying both HHV-6 and a cellular (host) housekeeping gene, demonstrated only in a proof-of-concept study, it could eventually
calculating the ratio of virus to the cellular housekeeping gene. be used for rapid phenotypic antimicrobial susceptibility testing.
Ratios of 1 HHV-6 copy to 2 cellular gene copies (as the cells are dPCR has also been used in the detection of parasites. In one study
diploid) were consistent with HHV-6 integration [20]. This assay using both genus- and species-specific assays, dPCR was used to
has been used to show reactivation of HHV-6 in 25% of hema- detect Plasmodium species from whole blood, demonstrating a high
topoietic stem cell recipients with donor- or recipient-derived sensitivity of 11 parasites/ml of blood [27]. The inhibitor-resistant
chromosomal HHV-6 [21]. nature of dPCR also makes it ideal for stool parasite quantification,
Quantification assays based on dPCR have also been developed for as shown by its ability to detect Cryptosporidium oocytes directly
other parts of the clinical microbiology laboratory besides virology. from fecal samples [28]. Assays are currently being developed to
dPCR was used to detect Mycobacterium tuberculosis DNA in the detect Schistosoma japonicum in urine, saliva, and stool but have
plasma of people with pulmonary tuberculosis (Tb) [22]. Though thus far been tested only in a mouse model [29].
the assay was relatively insensitive (65%), it represented a novel Another application of dPCR that is important for clinical micro-
and less invasive option for the diagnosis of Tb. The partitioning biology laboratories is its use for the characterization and standard-
of dPCR also makes it ideal to detect low numbers of resistant bac- ization of reference materials. dPCR can provide accurate, absolute
teria and viruses among a large population of sensitive organisms. quantification of reference materials and other control materials
dPCR detected Legionella pneumophila with gyrA mutations, which that are later used for qPCR. Researchers from the U.S. National
mediate fluoroquinolone resistance, at a ratio of 1 mutated allele Institute of Standards and Technology used dPCR on the BioMark
to 1,000 wild-type alleles [23]. Similar ratios of mutant to wild- HD instruments (Fluidigm) to quantify CMV DNA concentra-
type alleles were identified in a study that investigated the use of tion of the CMV standard reference material (SRM2366) they
dPCR to detect hetero-resistant Mycobacterium tuberculosis [24]. In produce for use in calibration of CMV quantitative assays [30].
that study, dPCR was more sensitive than both qPCR and Sanger Similarly, dPCR was used to quantify plasmid DNA standards for
sequencing, which could detect resistance only at 1:10 and 1:1 Shiga toxin-producing Escherichia coli with accuracy similar to that
mutant–to–wild-type allele ratios, respectively. Another reported of ultraviolet spectrophotometry and high-resolution inductively
application of dPCR is the detection of single-nucleotide poly- coupled plasma mass spectrometry [31]. Though the required dilu-
morphisms associated with antiviral resistance in influenza A virus, tion and fragmentation steps resulted in higher uncertainty in the
found in only 0.1% of the population tested, making dPCR 50 quantifications for dPCR, the method is much more accessible for
times more sensitive than the currently available clinical test [25]. laboratories that need to make their own control material.

Table 2. dPCR instruments and characteristics

Instruments
Partition No. of
Manufacturer type partitions Partitioning Amplification Detection Detection channels
Bio-Rad Droplet 20,000 QX200 droplet Thermal cycler QX200 droplet FAM/EvaGreen, VIC/HEX
Laboratories generator reader
RainDance Droplet 10,000,000 RainDrop source Thermal cycler RainDrop sense FAM/EvaGreen, VIC
Technologiesa

Life Technologies Chip 20,000 QuantStudio Dual flat block QuantStudio FAM/SYBR green, VIC,
(ThermoFisher) 3D chip loader thermocycler 3D reader ROX
and sealer
Fluidigm Chip 37,000 Biomark HD Thermocycler EP1 reader 2; FAM, VIC

Stilla Chip 30,000 Naica Geode Naica Geode Naica Prism 3 3; FAM, Cy3/VIC/Hex,
Cy5/Quasar 705
Formulatrix Chip 8,000- Constellation Constellation Constellation 5; FAM, VIC/HEX, and
12,000 digital PCR system digital PCR system digital PCR system user defined
jnmedsys Chip in 10,000 Clarity autoloader Thermocycler Clarity reader 2: FAM, VIC/HEX
a tube and sealer
a
Acquired by Bio-Rad Laboratories in 2017.

30 Clinical Microbiology Newsletter 40:4,2018 | ©2018 Elsevier


dPCR has also been used to characterize commercially available [7] Sedlak RH, Kuypers J, Jerome KR. A multiplexed droplet digital
reference materials. Three commercial standards for CMV quan- PCR assay performs better than qPCR on inhibition prone samples.
Diagn Microbiol Infect Dis 2014;80:285-6.
titation were tested using dPCR and qPCR, and both methods
[8] Jones GM, Busby E, Garson JA, Grant PR, Nastouli E, Devonshire
demonstrated deviations from the reference CMV concentration
AS, et al. Digital PCR dynamic range is approaching that of real-time
[32]. The two dPCR systems used showed no significant difference quantitative PCR. Biomol Detect Quant 2016;10:31-3.
in their quantifications, while the results from the qPCR systems [9] Pavsic J, Devonshire A, Blejec A, Foy CA, Van Heuverswyn F, Jones
varied with the assay utilized. dPCR was also used to characterize GM, et al. Inter-laboratory assessment of different digital PCR
the BK virus standards from the WHO and commercial provid- platforms for quantification of human cytomegalovirus DNA. Anal
ers [33]. While there was good agreement with the reference viral Bioanal Chem 2017;409:2601-14.
load for all providers, the WHO standard had a sub-population [10] Hayden RT, Gu Z, Sam SS, Sun Y, Tang L, Pounds S, et al. Compar-
ative performance of reagents and platforms for quantitation of cyto-
of viruses with deletions in the T region that could lead to under-
megalovirus DNA by digital PCR. J Clin Microbiol 2016;54:2602-8.
quantitation for PCR designed to target this region of the genome.
[11] Trypsteen W, Kiselinova M, Vandekerckhove L, De Spiegelaere W.
Similarly, dPCR was used to confirm next-generation sequencing Diagnostic utility of droplet digital PCR for HIV reservoir quanti-
findings that the WHO standard for JC polyomavirus had 4- to fication. J Virus Erad 2016;2:162-9.
8-fold variation in the copy numbers of different genes, which [12] Aizawa Y, Koyama A, Ishihara T, Onodera O, Saitoh A. Performance
would result in large variations in viral load in assays targeting of a real-time PCR-based approach and droplet digital PCR in
different regions of the genome [34]. detecting human parechovirus type 3 RNA. J Clin Virol 2016;84:27-
31.
Conclusions [13] Tang H, Cai Q, Li H, Hu P. Comparison of droplet digital PCR to
real-time PCR for quantification of hepatitis B virus DNA. Biosci
Although real-time PCR is a mature and well-established meth- Biotechnol Biochem 2016:1-6.
odology in clinical laboratories, some of its shortcomings have [14] Yan Y, Jia XJ, Wang HH, Fu XF, Ji JM, He PY, et al. Dynamic
increased the recent interest in dPCR. The potential applications quantification of avian influenza H7N9(A) virus in a human infection
of dPCR in the clinical microbiology laboratory cover a broad during clinical treatment using droplet digital PCR. J Virol Methods
2016;234:22-7.
range of purposes, targets, and specimen types. The applications
are likely to continue to expand dramatically as the availability and [15] Giovannelli I, Ciccone N, Vaggelli G, Malva ND, Torricelli F,
Rossolini GM, et al. Utility of droplet digital PCR for the quantita-
use of dPCR increases. Furthermore, variations of dPCR, includ- tive detection of polyomavirus JC in clinical samples. J Clin Virol.
ing digital loop-mediated isothermal amplification, have been pub- 2016;82:70-5.
lished (e.g. H5N1 quantification), with sensitivity comparable to [16] Jeannot E, Becette V, Campitelli M, Calmejane MA, Lappartient E,
that of qPCR and dPCR [35]. As the technology is used more in Ruff E, et al. Circulating human papillomavirus DNA detected using
clinical laboratories, variations of the amplification method used droplet digital PCR in the serum of patients diagnosed with early
will likely occur. For dPCR to supplant qPCR as the method of stage human papillomavirus-associated invasive carcinoma. J Pathol
Clin Res. 2016;2:201-9.
choice for quantification in clinical laboratories, several factors
[17] Gianella S, Chaillon A, Mutlu EA, Engen PA, Voigt RM, Keshavar-
(cost, workflow, and batch capacity) will need to be addressed and zian A, et al. Effect of CMV and EBV replication on intestinal muco-
optimized. More immediately, dPCR can be used as an additional sal gene expression and microbiome composition of HIV-infected
tool in the molecular arsenal. As molecular methods continue to and uninfected individuals. AIDS 2017; doi: 10.1089/aid.2017.0145.
develop at a rapid pace, dPCR will be an important tool in the [18] Arvia R, Sollai M, Pierucci F, Urso C, Massi D, Zakrzewska K.
clinical microbiology laboratory. Droplet digital PCR (ddPCR) vs quantitative real-time PCR (qPCR)
approach for detection and quantification of Merkel cell polyoma-
References virus (MCPyV) DNA in formalin fixed paraffin embedded (FFPE)
cutaneous biopsies. J Virol Method. 2017;246:15-20.
[1] Li HH, Gyllensten UB, Cui XF, Saiki RK, Erlich HA, Arnheim N. [19] Pavsic J, Zel J, Milavec M. Digital PCR for direct quantification of
Amplification and analysis of DNA sequences in single human sperm viruses without DNA extraction. Anal Bioanal Chem 2016;408:67-75.
and diploid cells. Nature 1988;335:414-7.
[20] Sedlak RH, Cook L, Huang ML, Magaret A, Zerr DM, Boeckh M,
[2] Saiki RK, Gelfand DH, Stoffel S, Scharf SJ, Higuchi R, Horn GT, et al. Identification of chromosomally integrated human herpesvirus
et al. Primer-directed enzymatic amplification of DNA with a ther- 6 by droplet digital PCR. Clin Chem 2014;60:765-72.
mostable DNA polymerase. Science 1988;239:487-91. [21] Sedlak RH, Hill JA, Nguyen T, Cho M, Levin G, Cook L, et al.
[3] Morley AA. Digital PCR: a brief history. Biomol Detect Quant Detection of human herpesvirus 6B (HHV-6B) reactivation in
2014;1:1-2. hematopoietic cell transplant recipients with inherited chromosom-
ally integrated HHV-6A by droplet digital PCR. J Clin Microbiol
[4] Vogelstein B, Kinzler KW. Digital PCR. Proc Natl Acad Sci 2016;54:1223-7.
1999;96:9236-41.
[22] Ushio R, Yamamoto M, Nakashima K, Watanabe H, Nagai K,
[5] Sedlak RH, Nguyen T, Palileo I, Jerome KR, Kuypers J. Superiority Shibata Y, et al. Digital PCR assay detection of circulating Mycobac-
of digital reverse rranscription-PCR (RT-PCR) over real-time RT- terium tuberculosis DNA in pulmonary tuberculosis patient plasma.
PCR for quantitation of highly divergent human rhinoviruses. J Clin Tuberculosis 2016;99:47-53.
Microbiol 2017;55:442-9.
[23] Hennebique A, Bidart M, Jarraud S, Beraud L, Schwebel C, Maurin
[6] Dingle TC, Sedlak RH, Cook L, Jerome KR. Tolerance of droplet- M, et al. Digital PCR for detection and quantification of fluoro-
digital PCR vs real-time quantitative PCR to inhibitory substances. quinolone resistance in Legionella pneumophila. Antimicrob Agents
Clin Chem 2013;59:1670-2. Chemother 2017;61:e00628-17.

Clinical Microbiology Newsletter 40:4,2018 | ©2018 Elsevier 31


[24] Pholwat S, Stroup S, Foongladda S, Houpt E. Digital PCR to detect [30] Haynes RJ, Kline MC, Toman B, Scott C, Wallace P, Butler JM,
and quantify heteroresistance in drug resistant Mycobacterium tuber- et al. Standard reference material 2366 for measurement of human
culosis. PLoS One 2013;8:e57238. cytomegalovirus DNA. J Mol Diagn 2013;15:177-85.
[25] Whale AS, Bushell CA, Grant PR, Cowen S, Gutierrez-Aguirre I, [31] Liang W, Xu L, Sui Z, Li Y, Li L, Wen Y, et al. Quantification
O’Sullivan DM, et al. Detection of rare drug resistance mutations by of plasmid DNA reference materials for Shiga toxin-producing
digital PCR in a human influenza A virus model system and clinical Escherichia coli based on UV, HR-ICP-MS and digital PCR. Chem
samples. J Clin Microbiol 2016;54:392-400. Central J 2016;10:55.
[26] Schoepp NG, Khorosheva EM, Schlappi TS, Curtis MS, Humphries [32] Hayden RT, Gu Z, Sam SS, Sun Y, Tang L, Pounds S, et al. Com-
RM, Hindler JA, et al. Digital quantification of DNA replication and parative evaluation of three commercial quantitative cytomegalovirus
chromosome segregation enables determination of antimicrobial standards by use of digital and real-time PCR. J Clin Microbiol
susceptibility after only 15 minutes of antibiotic exposure. Angew 2015;53:1500-5.
Chem 2016;55:9557-61.
[27] Srisutham S, Saralamba N, Malleret B, Renia L, Dondorp AM, [33] Bateman AC, Greninger AL, Atienza EE, Limaye AP, Jerome KR,
Imwong M. Four human Plasmodium species quantification using Cook L. Quantification of BK virus standards by quantitative real-
droplet digital PCR. PLoS One 2017;12:e0175771. time PCR and droplet digital PCR is confounded by multiple virus
populations in the WHO BKV international standard. Clin Chem
[28] Yang R, Paparini A, Monis P, Ryan U. Comparison of next-gener- 2017;63:761-9.
ation droplet digital PCR (ddPCR) with quantitative PCR (qPCR)
for enumeration of Cryptosporidium oocysts in faecal samples. Int J [34] Greninger AL, Bateman AC, Atienza EE, Wendt S, Makhsous N,
Parasitol 2014;44:1105-13. Jerome KR, et al. Copy number heterogeneity of JC virus standards.
J Clin Microbiol 2017;55:824-31.
[29] Weerakoon KG, Gordon CA, Cai P, Gobert GN, Duke M, Wil-
liams GM, et al. A novel duplex ddPCR assay for the diagnosis of [35] Hu Y, Xu P, Luo J, He H, Du W. Absolute quantification of H5-sub-
Schistosomiasis japonica: proof of concept in an experimental mouse type avian influenza viruses using droplet digital loop-mediated
model. Parasitology 2017;144:1005-15. isothermal amplification. Anal Chem 2017;89:745-50.

32 Clinical Microbiology Newsletter 40:4,2018 | ©2018 Elsevier

Anda mungkin juga menyukai