A Review of Novel Technologies and Techniques Associated With Identification of Bloodstream Infection Etiologies PDF

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Expert Review of Molecular Diagnostics

ISSN: 1473-7159 (Print) 1744-8352 (Online) Journal homepage: http://www.tandfonline.com/loi/iero20

A review of novel technologies and techniques


associated with identification of bloodstream
infection etiologies and rapid antimicrobial
genotypic and quantitative phenotypic
determination

Stephen Poole, Stephen P Kidd & Kordo Saeed

To cite this article: Stephen Poole, Stephen P Kidd & Kordo Saeed (2018) A review of novel
technologies and techniques associated with identification of bloodstream infection etiologies
and rapid antimicrobial genotypic and quantitative phenotypic determination, Expert Review of
Molecular Diagnostics, 18:6, 543-555, DOI: 10.1080/14737159.2018.1480369

To link to this article: https://doi.org/10.1080/14737159.2018.1480369

Accepted author version posted online: 23


May 2018.
Published online: 07 Jun 2018.

Submit your article to this journal

Article views: 111

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=iero20
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS
2018, VOL. 18, NO. 6, 543–555
https://doi.org/10.1080/14737159.2018.1480369

REVIEW

A review of novel technologies and techniques associated with identification of


bloodstream infection etiologies and rapid antimicrobial genotypic and
quantitative phenotypic determination
Stephen Poolea*, Stephen P Kidda* and Kordo Saeeda,b
a
Hampshire Hospitals NHS Foundation Trust, Department of Microbiology, Basingstoke and Winchester, UK; bUniversity of Southampton, School of
medicine, Southampton, UK

ABSTRACT ARTICLE HISTORY


Introduction: The antimicrobial aspect of management of patients with blood stream infections (BSI) Received 15 March 2018
and sepsis is time critical. In an era of increasing antimicrobial resistance, rapid detection and identifica- Accepted 21 May 2018
tion of bacteria with antimicrobial susceptibility is crucial to direct therapy early in the course of illness. KEYWORDS
Molecular techniques offer a potential solution to this. Blood stream infections;
Areas covered: In the present review the authors have discussed a number of novel solutions utilizing a antimicrobial susceptibility;
variety of molecular techniques for pathogen detection, identification and antimicrobial susceptibility. rapid diagnostics; PCR;
The review is not designed to be an exhaustive literature review covering all diagnostic solutions ever Accelerate
developed, instead the authors have focused on what they have had experience using, evaluating or
currently view as new and exciting with potential to revolutionize BSI diagnosis.
The authors searched PubMed (Medline) and Google Scholar with terms: BSI, Bacteraemia,
Candidaemia, Diagnostics, AST, Rapid, AMR, Novel and Blood Culture. The authors attended recent
clinical microbiology technology congresses.
Expert commentary: There are multiple exciting novel technologies at differing stages of development
with potential to revolutionize diagnosis of BSI. More work is needed as well as a standardized
assessment of different platforms in order to better understand the clinical and financial impacts
these will have in clinical microbiology laboratories.

1. Introduction antimicrobials is an increasingly daunting task due to an


ever-changing landscape of resistance mechanisms respond-
Blood stream infections (BSI) have carried an enormous burden of
ing to an impossibly complex web of selective pressures.
mortality globally since the beginning of recorded history and
Multiple epidemiological studies have shown shifts over
continue to do so [1]. Current gold standards for identification
time in the pattern of organisms causing sepsis and this
and treatment established by the Surviving Sepsis Campaign
will likely to continue in the future [1]. This highlights a
appear to have made some progress toward reducing this burden
need for rapid diagnostic tests that can not only assist
[2–4] but there is still a great deal more to do.
pathogen detection/identification, but also provide data on
Rapid identification and treatment is essential to improve
antimicrobial susceptibility and resistance mechanisms.
outcomes of patients who develop sepsis and septic shock. In
The wish of every infection specialist is to have a Star
this critical period an association has been well described
Trek™ style handheld Tricorder that could be used on septic
between timely administration of antibiotics and survival [5]
patients providing the ability to identify invading pathogens,
leading to the recommendation that these agents should be
determine antimicrobial susceptibility patterns and identify a
given within 1 hour [6].
source of infection in real time. We are a little way off that at
Correct and timely antibiotic administration has implica-
present but technology is rapidly providing new tools in the
tions both for patient survival and on wider antimicrobial
clinical laboratory and at the patient bedside.
resistance. It is frustrating, therefore, that the current standard
Rapid microorganism identification in signal positive
in testing typically takes at least 24 hours and can take up to
blood cultures has been achieved using established mole-
5 days. By this time the window for effective intervention may
cular techniques (Figure 1) like polymerase chain reaction
have closed.
(PCR)[9], fluorescent in situ hybridization (FISH) [10,11] and
A key aspect to improving outcomes is the targeting of
Matrix Assisted Laser Desorption Ionisation Time Of Flight
empirical antimicrobials toward suspected causative organ-
Mass Spectrometry (MALDI-TOF MS). The BioFire Filmarray®
isms and likely susceptibility patterns, especially, in the con-
[12], SeeGene Magicplex™ Sepsis and Bruker Biotyper
text of BSI [7]. Inadequate empirical therapy has been
Sepsityper®[13] are good examples of these innovations.
strongly associated with higher mortality [8]. Blindly targeting

CONTACT Kordo Saeed kordosaeed@nhs.net Consultant Microbiologist, Hampshire Hospitals NHS Foundation Trust, Basingstoke and Winchester, UK
*
Joint first authors.
© 2018 Informa UK Limited, trading as Taylor & Francis Group
544 S. POOLE ET AL.

Molecular
Diagnosis of
BSI

Signal Positive
Whole Blood
BC

Nucleic Acid Post- Nucleic Acid Post-


Novel Hybridisation Novel
Amplification amplification Amplification amplification

PCR + Array
Biosensors
Real-time PCR PCR + Array NMR FISH Real-time PCR PCR +
VOC analysis
Sequencing

Figure 1. Molecular techniques for diagnosis of blood stream infection. BSI: Blood stream infection; BC: Blood culture; NMR: Nuclear magnetic resonance; PCR:
Polymerase chain reaction; VOC: Volatile organic compound: FISH: Fluorescent in situ hybridization.

Additionally, some of the traditional bench side rapid latex robust, and reliable assays are now in use with no loss of
agglutination tests have been updated for use with signal diagnostic performance versus historical versions.
positive blood cultures like the Wellcogen™ range from
Oxoid used in the rapid qualitative detection of antigens
from Group B Streptococci, Neisseria meningiditis A, B, C, 2.1. AdvanDx – QuickFISH®
W135, Y, Haemophilius influenzae b, Escherichia coli K1. The QuickFISH® is the latest PNA-FISH (Peptide nucleic acid)
Currently there are a number of novel diagnostic tests iteration from AdvanDx and can report a result within 30 min
and technologies at the disposal of clinical microbiologists using a portfolio of four assays. It uses PNA probes targeting
and diagnostic laboratories that can support clinicians in bacterial 16S rRNA genes to directly identify organisms. The
better decision-making around diagnosis of infection staphylococcal assay identifies and differentiates
including BSI and antimicrobial stewardship. To our knowl- Staphylococcus aureus from coagulase-negative staphylococci
edge this is the most up-to-date and broad ranging review with a sensitivity/specificity of 97.1–100%/89.5–100%, well
regarding these techniques and technologies (Table 1) beyond that of tube coagulase tests [21,22] This technology
which adds novel developments and ideas to previous has also been applied in combination with the mecA
work in the field, as well as our own local views based XpressFISH® probe to rapidly identify methicillin resistant S.
on experience with several of these technologies [14–16]. aureus (MRSA) direct from signal positive blood culture [23].
We appreciate other diagnostic approaches are being Assays are also available for Enterococcus species with a sensi-
explored, in particular the field of differential gene expres- tivity/specificity 97.0–98.3%/100% [24], and the Gram-negative
sion profiles in sepsis [17,18]. The analysis of the host bacteria E. coli, Klebsiella pneumoniae and Pseudomonas aeru-
immune response to infection will help to identify unique ginosa with sensitivities for each organism of 98.4–100%,
biomarker signatures that could have diagnostic utility 95–99.9%, and 85.8–99.5%, respectively, with an overall speci-
when combined with pathogen detection strategies ficity of 94.8–99.6% [25]. Additionally there is a Candida spe-
[19,20]. This is an exciting field but does not fall within cies assay with sensitivity/specificity 98.3–100%/99.4–100%
the scope of this article. [26]. These assays are generally cheaper and require less
time to run than current real-time PCR assays.
Investigation into clinical applications has demonstrated a
significant improvement of patient outcomes, better antimi-
2. Fluorescent in situ hybridization (FISH)
crobial stewardship and reduced length of stay between those
FISH is not dependent on amplification of pathogen mate- receiving rapid identification and those not [27–29]. However,
rial making it less prone to inhibition and contamination a study by Holtzman et al found no benefit to the instigation
anomalies from the blood matrix and DNA from dead of staphylococcal PNA FISH without accompanying active
microorganisms. Historically FISH was quite laborious and reporting of results and antimicrobial stewardship advice [30]
time consuming; however rapid versions of these simple, reflecting findings with other rapid molecular tests[31].
Table 1. Summary of molecular methods for diagnosis of BSI discussed in this paper.
Tested
Technology Principle matrix Targets Turnaround time Hands-on time Advantages Disadvantages
AdvanDx QuickFISH ® FISH BC (1) GN – 3 (E.coli, K. pneumoniae, P. aeruginosa) <30 min <5 min Cheaper than PCR Limited number of probes
(2) GP – 2 (CoNS, S. aureus) No amplification needed, No AST
(3) Enterococcal – E. faecalis, E. faecium or other less contamination
enterococcus
(4) FP – 3 C. albicans, C. parapsilosis, C. glabrata
Accelerate FISH and Morphokinetic BC GN – 8 (4 species) GP – 6 (4 species) 2 Candida spp. ID 90 min, 2 min Rapid phenotypic sensitivity Reliability in polymicrobial
PhenoTest ™ BC Cellular Analysis (C. albicans, C. glabrata) AST ~7 h Minimal hands on time infection not established
AST – MIC Broad panel
Not gram stain dependent
Xpert ® MRSA/SA BC Real-time PCR BC MSSA, MRSA (mecA/C) 62 min <1 min Minimal hands on time Expensive
Robust platform
BD MAX ™ MRSA Staph Real-time PCR BC MSSA, MRSA (mecA/C, MREJ) 2h <1 min More amenable to batch
SR/XT testing than GeneXpert
Cognitor ® Minus ETGA BC Registers nonspecific presence of bacteria/fungi *12 h after 12 h Not published Performed on signal negative Very labor intensive in current
BC incubation blood cultures form
VERIGENE® BC-GP + PCR + Microarray BC (1) GP – 13 (9 species), mecA, vanA/B 2.5 h 10 min Unreliable in polymicrobial
BC-GN (2) GN 9 – (5 species), CTX-M, IMP, KPC, NDM, OXA, VIM. infection
Biofire Filmarray® BC PCR + Microarray BC GP – 8 (5 species), GN – 11 (9 species.), 5 Candida spp. 1h 2 min Not gram stain dependent Limited microbial ID +
mecA, vanA/B, KPC resistance panel
ePLEX® BCID-GN + PCR + Microarray BC GN – 21 (17 species), CTX-M, KPC, NDM, OXA, VIM, IMP 1.5 h <2 min Fully automated
BCID-GP + BCID-FP + pan-GP, pan-Candida.
GP- 20 (13 species), mecA/C, vanA/B + pan-GN, pan-
Candida.
FP- 16 (13 species).
Curetis PCR + Microarray BC GP – 11 (9 species), GN – 15 (14 species), FP 8, 4h <2 min Largest panel which is not
Unyvero BCU ™ Mycobacterial spp., 16 resistance genes gram dependent
Master diagnóstica PCR + Microarray BC GP – 7, GN 10, Candida albicans, 20 resistance genes 3h Not reported Not gram stain dependent
sepsis flow chip Large resistance gene
panel
Fully automated platform
available
Bruker Sepsityper ® MALDI-ToF MS BC Database dependent 30 min 5 min
a
Lightcycler SeptiFast
® PCR + Microarray WB GP – 6 (4 species), GN – 10 (10 species), FP – 6 (6 6h Not reported Not culture dependent
species). Optional mecA.
a
SeeGene Magicplex ™ Endpoint & Real-time WB GP – 73, 3 resistance genes vanA/B, mecA 6h Not reported, 2.5 Not culture dependent Need separate instrument for
PCR GN – 12 (12 species), 6 fungal species h extraction extraction
T2Sepsis Solution™ NMR WB GP – 2 (2 species) + GN – 4 (4 species) on the same 3–5 h Not reported Not gram stain dependent
(T2Dx ) ® panel
FP (5 species)
Specific diagnostics VOC analysis BC Database dependent 5h Not reported Unknown Unknown
SpecID
a

Q-Linea ASTrID Padlock probe PCR + WB 10 Genus, 33 species, 11 resistance genes 4 h ID, 10 h AST Not reported No blood culture required:
® Microarray AST – MIC on whole bloodb
Suitable for POC use
Q-Linea ASTar™ BC AST – MIC 6h Not reported Requires independent
organism ID
PCR + Microarray WB Not published Not published Not published
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS

Qvella FAST™ ID
a
Does not require standard culture – see notes bas claimed by manufacturer, published data has undergone a 4 h “enrichment” culture prior to starting the assay. BC: Blood culture; WB: Whole blood; NMR: Nuclear magnetic
resonance; AST: Antimicrobial Susceptibility; MIC: Minimum inhibitory concentration; GP: Gram-positive; GN: Gram-negative; FP: Fungal panel; POC: Point of care; ETGA: Enzymatic template generation and amplification.
545
546 S. POOLE ET AL.

2.2. Accelerate Pheno™ System – PhenoTest™ BC Multiplexed assays are desirable diagnostic solutions
because multiple tests can be replaced by a single reaction
To have a dual rapid identification and phenotypic AST is a
which in turn reduces turnaround time and saves precious
diagnostic challenge and currently there is only one platform
patient specimen for further investigations. The post-amplifi-
that is both CE marked and FDA approved for in vitro diag-
cation stage analysis is achieved with microarrays which are
nostic use. The Accelerate Pheno™ System promises organism
less labor intensive and are conducive to automation, unlike
identification at 1.5 hours via gel-filtration, FISH, and generates
sequence analysis. Whereas broad-range assays can in theory
a quantitative phenotype a further 5.5 hours later via morpho-
detect the presence of any pathogen, multiplex assays are
kinetic cellular analysis [32,33]. A number of centers (including
limited by the size of the arrays typically 10–50 targets, but
ours) have evaluated the PhenoTest BC assay performance for
they still possess significantly more targets than pathogen
ID and AST versus our standard of care (SoC) and the results so
specific PCR assays. Multiplex assays also offer the ability to
far appear promising. From the studies published to date the
detect the presence of resistance genes which broad-range
Pheno™ System has a sensitivity/specificity of 88.7–100%/
assays cannot. When combining a procalcitonin (PCT) test
97.1–100, EA and CA of 94.3–97.9%/95.1–97.6% (Minor Error
and/or other suitable biomarkers with PCR diagnostics, clinical
(MiE) 0–4.8%, Major Error (ME) 0.3–2.3%, Very Major Error
utility of these assays greatly increases [43].
(VME) 0.5–1.0%) [34–37].
The rapid results that are generated by the Pheno™ System
versus SoC have seen a reported dramatic decrease in the 3.1. Laboratory validated assays
turnaround time (TaT) of results. Marschal et al reported a
Developing a molecular test can ensure you are specifically
mean decrease in ID reporting by 27.47 hours and mean
answering a local diagnostic need. The cost is often signifi-
decrease in AST by 40.39 hours versus SoC [35]. They also
cantly smaller than purchasing one from a vendor and ulti-
had success with a number of correctly reported E. coli
mately the control and target choice is yours. However, there
extended spectrum beta lactamases (ESßLs) and multi-drug
are a number of hurdles that have to be overcome to collect
resistant (MDR) P. aeruginosa. They also report that the
these benefits. Regulatory and quality requirements of a
Pheno™ System detected further pathogens that would other-
laboratory developed test (LDT) can be complex and time
wise have been reported as monomicrobial by SoC, this has
consuming and the initial expertise needed for development
been our experience as well.
is not always available in the clinical microbiology department.
To date there has been no published data relating to the
In the UK, Public Health England has developed a number of
improved stewardship or clinical outcomes of the Accelerate
algorithms and standards for microbiological investigations
Pheno™ System, but locally we have made a number of
(SMIs) including those involved in transitioning a LDT into
clinical decisions and antimicrobial interventions using the
clinical service [44].
system, especially in the context of resistant Gram-negative
Blood is a complex biological matrix and harbors many
pathogens. This has included de-escalation and patient speci-
components that are inhibitory to PCR including both haem
fic targeting of antimicrobial therapy in sepsis and reducing
(from lysed erythrocytes) and immunoglobulins. It is crucial
our burden of broad-spectrum antibiotics. Our manuscript for
the extraction, buffer and DNA polymerase utilized must be
the clinical impact analysis of the Pheno™ System is currently
robust enough to deliver an assay with reproducible perfor-
being prepared.
mance. The Phusion Blood Direct PCR Kit by ThermoFisher [45]
is a good example of such a development that could be
utilized by a LDT [46]. A second approach would be to validate
3. Nucleic acid amplification of signal positive blood a previously developed PCR for use on signal positive blood
culture – polymerase chain reaction (PCR) cultures that had not been initially indicated by the original
developers. This is an approach we at Hampshire Hospitals
The portfolio of diagnostic PCR strategies that are currently in (HHFT) are undertaking using the r-biopharm RIDA®GENE
use comprise pathogen specific, broad-range, and multiplex
MRSA [47] real-time PCR. This approach could open up avail-
assays [38]. The fundamental goal that each method shares is able diagnostic real-time PCR assays for use on signal positive
to amplify pathogen DNA against a background of host DNA.
blood cultures, but increased awareness of quality procedures
Pathogen specific assays employ genus/species level detection
is crucial to avoid issues with regulatory authorities.
for the likes of Candida spp. or MRSA but are traditionally
limited to <6 targets due to technical limitations of the ther-
mocycler optics. 3.2. Cepheid GeneXpert – Xpert® MRSA/SA BC
Universal broad-range assays amplify conserved regions of
The GeneXpert is a modular platform combining nucleic acid
the isolated microorganism; usually 16S/18S rRNA [39] or other
extraction and amplification that can be expanded to fit clin-
housekeeping genes which help broaden the scope of patho-
ical laboratory requirements [48]. This simple to answer solu-
gen detection. However, there is an associated post-amplifica-
tion provides the ability to detect pathogens within 1 hour
tion stage to complete the identification when using this
from sample preparation from numerous specimen types. The
technique that requires sequencing of amplicons [40];
Xpert® MRSA/SA BC real-time PCR assay requires minimal
Molzym have developed the SeptiTest™ [41,42] workflow as
hands-on-time, <5 mins, and can be operated by a non-labora-
an example. This approach can help to elucidate culture nega-
tory specialist directly on a signal positive blood culture fol-
tive/fastidious pathogens from negative blood cultures.
lowing an indicated Gram-stain [49–52]. It targets the Spa,
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS 547

SSSmec cassette and mecA genes with a sensitivity/specificity solutions discussed offer differing amounts of automation
of 100%/98.6% for S. aureus and 98.3%/99.4% for MRSA, from very separate distinct stages present in the VERIGENE®
respectively. The Xpert® MRSA/SA BC assay provides the infec- to the GenMark DX ePlex® fully automated sample-to-answer
tion specialist with a powerful tool for rapid antibiotic inter- solution.
vention in areas with high prevalence of MRSA bacteremia or a The use of sequence analysis as a post-amplification tool
de-escalation stewardship tool if MRSA antibiotics constitute has been achieved and offers a very broad-range of pathogen
part of empirical regimen. detection [14,32,42,67–69]. However, it is neither novel nor
rapid and only has a role in a reference laboratory in its
current format. Deurenberg et al review the potential future
3.3. BD MAX™ – StaphSR and the new MRSA XT assay
impact of next generation sequencing (NGS) in clinical micro-
The BD MAX™ System is a fully-integrated and automated biology and how advancements in assay design and data
platform that performs nucleic acid extraction and real-time analysis improvements could one day translate to appropriate
PCR with <5 min sample preparation time. The sensitivity and diagnostic assays for clinical microbiology [70].
specificity of the SR assay has been reported 100%/100% for S.
aureus, and 97.9%/98.1% and 99.0%/95.8% for MRSA and
4.1. Luminex nanosphere VERIGENE® – BC-GP & BC-GN
coagulase-negative staphylococci, respectively [53,54].
According to the manufacturers the new MRSA XT assay The VERIGENE® combines automated nucleic acid extraction
has been designed to detect the novel MREJ types [55,56] not and PCR amplification on the ProcessorSP followed by hybridi-
detected by other molecular assays as well as MRSA strains zation to Nanogrid arrays with gold nanoparticle detection.
with the novel mecC gene which can account for 3–4% of all The manufacturers claim that the gold nanoparticle is more
cases [57]. It can also detect mecA dropouts which can make sensitive than fluorophore detection. The platform work flow
up to 18% of all positive MRSA results reported by other requires a Gram-stain to select the appropriate assays and is
molecular assays which in fact are actually false positives not fully automated so would need to be placed within a
[58,59]. This platform lends itself to batch testing with its clinical laboratory [71].
medium throughput capacity compared to the GeneXpert This platform has a portfolio of different species and genus
and can be operated by junior laboratory staff thus lowering probe targets for both Gram-positive organisms (92–99%
costs. agreement with conventional methodology) [72–74] and
Gram-negative organisms [75–77]. The assay time is rapid,
taking approximately 10 min of hands on time followed by a
3.4. Momentum bioscience – Cognitor® Minus
2.5 hour run. The VERIGENE also detects common resistance
Cognitor® Minus uses enzymatic template generation and genes with a high level of accuracy.
amplification (ETGA) technology [60,61] that has been devel- At this time there is limited published data about the
oped to detect microorganisms by measuring nucleic acid clinical impact of these platforms. Stewardship reviews of the
modifying enzymes present within their cells [62,63]. This Gram-negative panel suggest optimization of antimicrobials
elegantly unique approach provides a rapid and sensitive can occur considerably earlier [75,78]. This is reflected in stu-
universal detection of all bacterial and fungal species present dies with the Gram-positive portfolio [72] albeit in a demo-
in blood culture on any standard thermocycler likely to be graphic of patients with high rates of MRSA, fortunately
already present in a standard clinical laboratory. It does not something which is less prevalent in our practice in the UK.
rely on a preselected panel of microorganisms so is free to In high resource settings there has been demonstrated
detect any viable pathogens present. Initial clinical evaluation improvement in length of ICU stay and 30 day mortality
performed locally by Dryden et al has shown that this early when using these platforms [77]. However a study of 168
“rule out” of a BSI with a negative predictive value of 99.5% episodes of bacteremia in the US found no significant change
versus 5 day negative blood cultures can be a powerful tool in in clinical outcomes (mortality and length of stay) when using
antimicrobial stewardship and patient management [64]. In its the Gram-positive platform compared to standard of care,
current form this test is quite laborious and requires specialist albeit with significantly improved time to initiation of appro-
laboratory staff; however a rapid automated version of this priate antibiotics with a 24 hour reduction [79]. This reflects
assay that can be used directly on whole blood (WB) without research on other rapid diagnostic platforms that suggest
the need for culture is imminent. rapid turnaround times are irrelevant if not coupled with
good, timely stewardship advice.
According to manufacturer’s information and published
4. Post-nucleic acid amplification detection –
data [72] these assays are unreliable in the detection of organ-
microarray and sequencing approaches
isms from polymicrobial cultures.
Microarrays offer the potential for a highly multiplexed diag-
nostic solution. They combine the utility of PCR amplification
4.2. BioMeriéux – Biofire® FilmArray®
and hybridization to oligonucleotide probe arrays of differing
sizes. The true microarrays were developed for the analysis of This platform provides a sample to answer solution with syn-
differential gene expression [65,66] with probes numbering dromic panels that do not require a Gram-stain. The ease of
tens of thousands, so in reality for the purist, the arrays used use and simple work flow lends itself to be used at point-of-
in diagnostics should be referred to as macroarrays. The care and out-of-hours with extraction, PCR amplification and
548 S. POOLE ET AL.

array hybridization occurring within a sealed pouch. The run 4.5. Master Diagnóstica – Sepsis Flow Chip assay
time is 60 min and requires <5 min of assay pouch preparation
The Sepsis Flow Chip (SFC) assay comprises a multiplex PCR
involving hydration of reagents and inoculation of signal posi-
and hybriSpot automated reverse hybridization to a low den-
tive blood culture [80].
sity DNA array [90]. It can detect seven Gram-positives (incl. S.
The targets on the blood culture panel comprise 6 Gram-
aureus, S. pyogenes, L. monocytogenes), 10 Gram-negatives
positive (incl. Listeria monocytogenes, Streptococcus pyogenes,
(incl. E. coli, P. aeruginosa, N. meningitidis) Candida albicans,
Streptococcus pneumoniae), 10 Gram-negative (incl. H. influen-
and 20 resistance genes. Galiana et al have done an early
zae, N. meningitidis, K. pneumoniae), and 5 Candida species,
evaluation of the assay reporting a sensitivity and specificity
plus mecA, vanA/B and KPC resistance genes. It is a well-
of 93.3%/100% for ID and 93.6%/100% for the resistance
established assay and platform with many evaluations and
genes [91]. They conclude that the system is user friendly
technology comparisons in the published literature [81–85]
and easy to interpret the results.
reporting sensitivity and specificity of 89.4–91.6%/100%. In
comparison to the other array based assays the panel is lim-
ited in size but has more variety; with the likes of N. meningi-
tidis and L. monocytogenes included that are not often present
5. MALDI time-of-flight mass spectrometry
on similar platforms but is lacking with the selection of resis-
tance genes offered. It has seen clinical evaluations where The use of MALDI-TOF MS for detection of bacteria and fungi
rapid ID has led to appropriate de-escalation of antibiotics isolated from patient clinical specimens has been well docu-
<14 hours faster [84]. However, having a full BC assay without mented and has revolutionized the work flow in clinical micro-
the need for Gram-stain driven selection of appropriate panels biology laboratories [92–96]. The most common MS solutions
is an advantage especially if the proposed use in outside of a in use are Bruker Daltonics Biotyper® (MBT) and bioMérieux
clinical microbiology laboratory like in ED/ITU or an acute Vitek® MS which are becoming the workhorses of many clin-
assessment ward (AAU). A new BCID panel is under develop- ical microbiology laboratories after recent universal adoption
ment and is undergoing beta-testing by Blaschke et al that [93]. The protein spectral databases of microorganism are
incorporates >25 microorganisms [86]. increasing for each MS solution and the identification that
MALDI-TOF MS provides is as good as currently available
biochemical and nucleic amplification tests [92].
4.3. GenMark DX – ePlex® BCID
The desire for rapid diagnostics on signal positive blood
This is a highly multiplexed fully automated one-step single- culture has seen attention focused on using MALDI-TOF MS.
use cartridge assay system that won a silver medal at the 2017 The use of this technique direct on blood is more challenging
Medical Design Excellence Awards [87]. Unlike the previous that on other less complex specimen types due to the complex
two platforms the reaction cassette only requires inoculation matrices contained within a blood culture bottle that interferes
and placing in the control module. There are three options with the generation of good quality spectra [97]. Numerous
available for signal positive blood cultures post-Gram stain: groups are publishing peer-reviewed studies of their own labora-
BCID-GP, BCID-GN and BCID-FP. The Gram-negative assay has tory developed methods to remove the need for traditional sub-
21 targets, 6 resistance genes and a pan-GP and pan-Candida culture. The two most common methods are either lysis/extrac-
target, the Gram-positive assay has 20 targets, 4 resistance tion methods on a prepared bacterial/fungal pellet [98,99] or
genes, pan-GN and pan-Candida target. The fungal assay has from a novel rapid <6 hours subculture approach using pre-
16 targets. There only appears to be one published review of warmed solid media first described by Idelevich et al [100–102].
the ePlex® system, and this details the BCID-FP assay [88]. Due to the time sensitive nature of a signal positive blood culture
Despite requiring a Gram-stain for selection of the correct report a direct subculture independent method is the most rapid
assay it does have the helpful feature of having pan-targets and desired approach. This method has been compared to post-
present to capture a pathogen from a misread Gram-stain or blood culture PCR assays with similar results at a fraction of the
an unrecognized polymicrobial blood culture. cost [85]. The sensitivity of this direct blood culture MALDI-TOF
MS analysis has been established in the region of 80–90%
[103,104]. It must be noted that if you delve into the finer details
4.4. Curetis Unyvero™ – BCU
of these data Gram-negative sensitivity and concordance at
The Unyvero™ modular concept provides a sample to answer genus/species level is much higher. The main issues MALDI-
solution in 4 hours using a cartridge that according to the manu- TOF MS has is distinguishing viridans streptococci from S. pneu-
facturer contains more components than any other on the market, moniae [105] which accounts for the Gram-positive scores
“eight labs on eight chips in a brick”. Targets include 10 Gram- becoming significantly lower. These problems are compounded
positive (including e.g. S. aureus, S. pyogenes), 15 Gram-negative by the background noise even the best sample preparation
(e.g. E coli, K. pneumoniae), Mycobacterium tuberculosis, eight fun- method leaves behind. The quantity of lower m/z peaks causes
gal, and 16 resistance genes. This is the largest panel currently on a lowering of the quality score because they are not present on
offer which can be used without the need for a Gram-stain making the corresponding microorganism spectra in the manufacturer’s
it potentially more suited to a point-of-care application outside of database. This problem is alleviated by the introduction of the
laboratory hours in ED/ITU or AAU. Currently clinical evaluations blood culture analysis software module on the Bruker Biotyper. It
are being undertaken in Germany and the US but there is no must be noted that the analysis of the generated protein spectra
published clinical application data at this time [89]. is only as good as the database that is interrogated.
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS 549

5.1. Bruker Biotyper – Sepsityper® kit recommended 5 days incubation to report a negative.
Sensitivity for acutely ill patients is ~70% and even lower for
This is essentially a sample processing kit to enable the stan-
fastidious microorganisms [116]. Analysis of patient whole
dardization of the sample preparation process associated with
blood without the need for blood culture has the potential
direct analysis of signal positive blood cultures. There are
to dramatically decrease the time to pathogen identification
numerous laboratory developed methods but this has led to
versus blood culture. These would be used on patients pre-
inconsistent sensitivity and specificities reported and most are
senting in the emergency department or symptomatic inpati-
no more than research tools. When compared to general
ents. The amplification of the pathogen signal during real-time
standard of care of sub-culture and classical phenotypic iden-
PCR is crucially important owing to the low numbers of circu-
tification methods the Sepsityper® kit generates a genus/spe-
lating bacteria, 10 CFUs/ml or fungi, 1–10 CFU/ml, reported in
cies concordance with SoC of 94.7–100%/94.7–100% for Gram-
adult sepsis. Direct molecular assays also facilitate detection of
negatives and 91.7–93.3%/70–73.1%- for Gram-positives when
fastidious, difficult-to-culture organisms such as fungi and
using the blood culture analysis software module [106].
common intracellular bacteria.
Morgenthaler et al have completed a comprehensive meta-
analysis of studies involving the Sepsityper® kit [107] which
makes good reading. They conclude having a standardized CE- 6.1. Roche LightCycler® – SeptiFast test
IVD marked kit for processing of signal positive blood cultures
There are many studies evaluating its diagnostic performance
does remove the regulatory requirements associated with
and clinical impact owing to the many years it has been
using a laboratory developed method despite the cost impli-
available [117–122]. However, in the UK it was the subject of
cations involved.
a recent Health Technology Assessment study [43] commis-
sioned by the UK’s National Institute for Health Research
5.2. MALDI-TOF MS and antimicrobial susceptibility (NIHR). The sensitivity and specificity from this study was
testing (AST) 50%/85.8%, which was lower than the pooled data from the
meta-analysis however this was a larger well planned multi-
It is not surprising that the use of MALDI-TOF MS is being center prospective evaluation than those previously under-
explored for the generation of a phenotypic AST. We have taken. As a result its recommendation was the SeptiFast was
discussed the positive impact it has had in routine clinical not suitable to replace blood culture, but could be used to
microbiology and now more so in processing of signal posi- augment current SoC in place within clinical microbiology. A
tive blood cultures. The majority of the methods that are positive result could be acted upon without waiting for blood
being developed are very much in their infancy and are culture to signal positive but a negative result did not indicate
presented as research tools but represent some exciting an absence of pathogen.
uses of MALDI-TOF MS. DeMarco et al, Faron et aland Vella
et al provide well written reviews on the potential
approaches the MALDI-TOF MS could use in generating a 6.2. SeeGene – Magicplex™ sepsis
phenotypic AST [108–110]. There are four avenues in devel- SeeGene have created a full process for analysis of whole
opment that appear to be the most popular [92] (i) Detection blood incorporating pathogen DNA extraction with the
of the modification of an antibiotic i.e. the hydrolysis product SelectNA blood pathogen kit automated on the SEEPrep12.
of a ß-lactam demonstrated by the MBT-STAR assay [111] and The extracted DNA is amplified using the novel combination
the newly CE-IVD marked MBT-STAR Carba assay from Bruker of endpoint PCR and real-time PCR assay that offers a highly
for rapid detection of Class A, B or D carbapenemase activity multiplexed assay without the need for blood culture. The
in Gram-negatives [112] (ii) Detecting the evidence of meta- pathogen identification and possible presence of resistance
bolism of a pathogen in the presence of an antibiotic, dead genes is reported at 6 hours. The panel consists of 73 Gram-
(S) or alive (R), MBT-RESIST assay [113], (iii) Detection of an positives, 12 Gram-negatives, mecA, vanA/B and six fungi,
antibiotic action on an antibiotic target site [114], (iv) Semi- providing 90% coverage of sepsis related pathogens. An
quantification of the presence of intracellular or extracellular early studies show that it can applied in hospital adult and a
antibiotic, the MBT-ASTRA assay has been shown to produce pediatric populations but as yet there are no further published
equivalent MICs to ETEST® MICs on the antibiotics that were clinical evaluations [123,124].
tested by Sparbier et al [115].
Whichever further methods progress through standardiza-
tion and clinical evaluation they would need to be comparable 6.3. T2 biosystems – T2Dx® T2Candida® & T2Bacteria®
to the ‘gold standard’ broth microdilution (BMD) method to The T2Dx platform is a novel diagnostic technology that uti-
pass the regulatory requirements set out by EUCAST and CLSI lizes nuclear magnetic resonance as the detection mechanism
like the MBT-STAR Carba assay. with a run time of 3 hours [125]. Two assays are available on
the T2Dx: T2Bacteria®, and T2Candida®. The T2Bacteria® iden-
tifies six bacterial species (E. coli, K. pneumoniae, P. aeruginosa,
6. Direct – blood culture independent (whole blood)
Acinetobacter baumannii, S. aureus, and Enterococcus faecium)
Traditional blood culture processing ‘the gold standard’ is with at least two of these often not covered by empirical
widely used in routine diagnostic laboratories, however patho- therapy. The T2Candida® identifies five Candida species (C.
gen identification can take 24–48 hours or even longer, with albicans, Candida parasilosis, Candida krusei, Candida tropicalis
550 S. POOLE ET AL.

and Candida glabrata). Together both panels cover 90% of VOCs via a colorimetric array [142,143]. Full clinical diagnostic
Gram-negative and 70% community acquired infections performance has yet to be established but this represents an
encountered in the emergency department (ED) [126] where exciting development in clinical laboratory medicine.
we believe this would be the perfect placement of the system.
At this time only the T2Candida® has been the subject of
7.2. Q-Linea- ASTrID® & ASTar™
analytical and clinical evaluations [127–130]. The large pro-
spective study involving the T2Candida® conducted by Q-Linea are offering two novel diagnostic and quantitative
Clancy et al [131] reported a clinical sensitivity of 89% com- AST phenotypic solutions, one from whole blood, ASTrID®
pared to blood culture and that T2 positivity was associated and one from signal positive blood cultures, ASTar™. The
with current antifungal therapy, neutropenia and C. albicans ASTrID® generates pathogen identification using highly spe-
BSI. The T2Bacteria® assay has yet to have any performance cific and selective padlock probes [144,145] forming circular-
data published but having both assays in use outside of the ized DNA strands[146] that are amplified via rolling-circle
clinical microbiology laboratory in ED/ITU operated by non- amplification (RCA) [147] and subsequent circle-to-circle
specialized staff on whole blood would dramatically decrease amplification (C2CA) [148]. The amplified products are fluores-
diagnostic uncertainty and help facilitate rapid targeted cently labelled and detected on a microarray. A poster at the
treatment. ASM 2017 highlighted an evaluation of the ASTrID® by Morck
et al in Sweden [149]. Identification was performed on 4 hours
incubated blood cultures. Sensitivity and specificity versus
7. Novel diagnostic solutions and future directions
blood cultures taken at the same time from patients attending
When reading published literature there are many novel and an infectious disease ED in Örebro, Sweden was 97%/99.6%.
ingenious proposed solutions to improve diagnostics of BSI. The AST was performed on signal positive spiked blood cul-
For example, Templer et al describe a proof of concept micro- tures and reported a 96% essential agreement (EA) and 95%
array coupled with a Surface Plasmon Resonance Imager (SPRi) categorical agreement (CA) compared to reference broth
to detect bacteria in clinical specimens [132] with a limit of microdilution (BMD).
detection of 1 CFU/ml of blood. This would make it suitable for
detection of a BSI on whole blood without the need for
7.3. Qvella – FAST™ ID
culture. Another on the horizon is the utilty of NGS within
clinical microbiology, moving out of the reference laboratory This is a novel way of addressing the sensitivity issues surround-
and closer to the patient. However, there are a number of ing direct PCR on whole blood. Qvella has developed the FAST™
hurdles to overcome first like the adoption of a universal ID technology to rapidly concentrate bloodstream pathogens
library preparation method to facilitate standardization and and release their nucleic acids electrically. While groups focus
creation of a user friendly data analysis interface for a start. on developing more sensitive detection methods to enhance the
Resources will undoubtedly be directed to streamline NGS performance of their assays, Qvella has focused on optimizing
where the unbiased nature of metagenomic analysis will sample preparation via automation of isolation, concentration
enhance the breadth of pathogen detection, especially help- and lysis of a small number of pathogens in a whole blood
ing in cases where conventional diagnostics have not suc- sample. This unique approach is designed to achieve rapid and
ceeded [133,134]. We have seen targeted NGS used in near automated pathogen identification in minutes, not hours, and is
real-time during the Ebolavirus pandemic 2014–2016 when expected to play a key role in enabling earlier tailored antimicro-
the Oxford Nanopore MinION was used in the field to bial treatment. This system is currently not available to purchase
sequence clinical specimens for Ebolavirus surveillance toward but could provide an exciting diagnostic option if placed in the
the end of the outbreak with great success [135]. emergency department to help stratify patient management and
Many of these novel approaches are not yet close to com- flow [150].
mercial development and therefore not ready for a routine
clinical diagnostic use. Here we discuss a few exciting plat-
8. Expert commentary
forms that have made it to early analytical and clinical
evaluation. Here we present a number of novel technologies and techniques
that are either available or close to market. Clinical microbiology
aims to provide rapid and accurate diagnostics to clinicians to
7.1. Headspace volatile organic compound (VOC)
improve patient outcome. This is particularly relevant in BSI in
analysis – SpecID™
order to assist targeted antibiotic therapy and reduce the use of
As bacteria and fungi respire in blood culture they inevitably unnecessary broad-spectrum antibiotics.
emit the waste products of this process as they divide and In light of the pressures associated with continual improve-
multiply [136–138]. The headspace present in the blood culture ment within financially challenged healthcare systems it is
bottle has been analyzed [139] for unique signatures of these vital each new iteration of a diagnostics test be faster, easier
microorganisms and data has shown that this signal can be to use, more sensitive/specific and affordable. If it cannot
diagnostic [140,141]. This technology has been adapted by provide all of these features it must at least have a comparable
Specific Technologies to a laboratory based diagnostic test diagnostic performance, be more rapid and demonstrate an
called SpecID™ that promises pathogen identification from sig- economical benefit otherwise laboratories will have major
nal positive blood culture in 5 hours. It is based on detection of challenges to adopt them. This has been a particular hurdle
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS 551

in the adoption of molecular assays with are generally con- aimed to complement clinical decision-making at the patients’
siderably more expensive than traditional methods. Culbreath bed side. These devices do not require operation by laboratory
et al [151] propose a six-step model for test adoption that professionals, but by staff that are trained and in direct con-
appraises a whole process approach from ordering of the test tact with the patients. We see molecular testing moving more
to reporting of the actionable result and shows much merit. toward near-point testing where clinical decisions can be
The performance of blood culture as a diagnostic test is influenced in real time.
made up of multiple components extending well beyond the All of the reviewed molecular tests dramatically improve the
diagnostic assays including pre-analytical (arm to incubation), turnaround time of results with high levels of sensitivity and
analytical (incubation to result) and post-analytical (commu- specificity versus traditional culture methods. In spite of this,
nication of result) stages. It is important in an era of revolu- evidence showing improvement in clinical outcomes is sparse
tionary molecular diagnostics in BSI that we do not lose sight and there is evidence that this impact is minimized without
of the need for robust pre- and post-analytical systems. The adequate support of clinicians from antimicrobial stewardship
UK SMI [152] recommended a 4-hour target for incubating teams. Moving forward infection specialists must develop robust
collected blood cultures which is often breached out of nor- systems to communicate the significance of results to clinical
mal working hours. Optimization of the pre-analytical stage teams so that the impact can be maximized. Further studies are
has been shown to improve turnaround time [153,154]. This needed to shed more light on the effectiveness of these tests,
can be achieved by either having a 24 hour service in the both in financial terms and clinical outcomes. We hope collea-
hospital with a pan-pathology approach [155] or by placing a gues who work on these technologies and or evaluate them
satellite incubator in ED or ITU [156], especially effective at clinically are able to publish and share these data as we aim to
sites that operate a “hub and spoke” clinical laboratory system. do with our Accelerate Pheno™ System clinical evaluation.
Quality is an integral part of clinical microbiology and numer- Direct testing of whole blood is the most exciting develop-
ous schemes are in place to ensure a fit for purpose service is ment on the horizon. If reliable pathogen identification and a
being delivered. External quality schemes (EQA) are one of the quantitative phenotype AST is achieved this will have an
tools employed in this process. However, the EQA schemes can be undoubted impact in patient outcomes in sepsis. Currently
slow to adapt to novel technologies and so this important quality developments have not replaced blood culture but will aug-
tool is not always available. Local laboratories could initiate inter- ment the portfolio of tests at the infection specialist disposal
lab exchange schemes in lieu of an EQA and offer to collaborate to diagnose and treat BSI. The reduction in diagnostic uncer-
with national schemes and manufacturers to set one up. tainty in sepsis and BSI is the key for all.
During the review preparation it has become apparent that the
standard of the evaluations of new assays and platforms are of
Key issues
differing quality. This makes it problematic to compare the studies
to present fair comparisons. Despite the Standards for Reporting ● There is an unmet need to reduce diagnostic uncertainty in
of Diagnostic Accuracy Studies (STARD) guideline being updated sepsis and BSI.
in 2015 to harmonize the reporting of such studies there appears a ● The use of novel molecular technologies offers solutions to
lack of uptake. The guidelines were written to allow the readers to augment patient care and reduce mortality.
judge the trustworthiness and applicability of study findings [157] ● The increased speed and sensitivity offered by this technol-
and to facilitate easier comparisons. Blood culture is universally ogy requires robust post-analytical systems in order for
accepted to be insensitive which can be affected by the presence clinical and economic impacts to be maximized.
of fastidious organisms or antibiotic pre-treatment. Novel mole- ● There is a need for higher quality, standardized appraisal
cular tests are designed to be more sensitive, herein lies a problem studies evaluating new diagnostic technologies.
for designing a robust technology evaluation. The published stu- ● Novel diagnostics direct on whole blood is becoming an
dies we have reviewed have often reported a molecular test as exciting field.
positive versus a negative blood culture. These are difficult to
interpret, where dead bacterial DNA may be muddying the waters.
Funding
The increased quality standard would encourage better designed
studies where the imperfect “gold standard” comparator of blood This paper was not funded.
culture could be augmented with biomarkers and clinical details
and a third test, like sequencing would be used to help interpret
Declaration of Interest
the discrepant results. We hope the diagnostic community uni-
versally adopts STARD to ensure future research and technology The authors have no relevant affiliations or financial involvement with any
evaluations are of a higher quality. organization or entity with a financial interest in or financial conflict with
the subject matter or materials discussed in the manuscript. This includes
employment, consultancies, honoraria, stock ownership or options, expert
testimony, grants or patents received or pending, or royalties. Peer
9. Five-year view
reviewers on this manuscript have no relevant financial or other relation-
The landscape of traditional diagnostic laboratories is chan- ships to disclose.
1. Martin GS, Mannino DM, Eaton S, Moss M. The epidemiology of
ging; there is a major emphasis in the UK and elsewhere in the
sepsis in the United States from 1979 through 2000. N Engl J Med.
developed world on laboratory mergers and consolidations. 2003;348:1546–1554.
Parallel to this there is advance in diagnostic technology, 2. Paul, M., Shani V, Muchtar E, Kariv G, Robenshtok E, Leibovici L .
devices and innovative point-of-care products which are Systematic review and meta-analysis of the efficacy of appropriate
552 S. POOLE ET AL.

empiric antibiotic therapy for sepsis. Antimicrob Agents performed on positive blood cultures for evaluation. J Clin
Chemother. 2010;54:4851–4863. Microbiol. 2013;51:131–135.
3. Zimmerman JE, Kramer AA, Knaus WA. Changes in hospital mortal- 23. Salimnia H, Fairfax MR, Lephart P, et al. An international, prospec-
ity for United States intensive care unit admissions from 1988 to tive, multicenter evaluation of the combination of AdvanDx
2012. Crit Care. 2013;17:R81. Staphylococcus QuickFISH BC with mecA XpressFISH for detection
4. Kaukonen K-M, Bailey M, Suzuki S, Pilcher D. Mortality related to of methicillin resistant Staphylococcus aureus isolates from positive
severe sepsis and septic shock among critically ill patients in blood cultures. J Clin Microbiol. 2014;52:3928–3932.
Australia and New Zealand, 2000-2012. JAMA. 2014;311:1308. 24. Deck MK, Anderson ES, Buckner RJ, et al. Rapid detection of
5. Kumar A, Roberts D, Wood KE, et al. Duration of hypotension Enterococcus spp. direct from blood culture bottles using Enterococcus
before initiation of effective antimicrobial therapy is the critical QuickFISH method: a multicenter investigation. Diagn Microbiol Infect
determinant of survival in human septic shock. Crit Care Med. Dis. 2018;78:338–342.
2006;34:1589–1596. 25. Della-Latta P, Salimnia H, Painter T, et al. Identification of
6. Rhodes A, Evans LE, Alhazzani W, Levy MM, et al. Surviving sepsis Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aerugi-
campaign: international guidelines for management of sepsis and nosa in blood cultures: a multicenter performance evaluation of a
septic shock: 2016. Intensive Care Med. 2017;43:304–377. three-color peptide nucleic acid fluorescence in situ hybridization
7. Kreger BE, Craven DE, McCabe WR. Gram-negative bacteremia. Am assay. J Clin Microbiol. 2011;49:2259–2261.
J Med. 1980;68:344–355. 26. Abdelhamed AM, Zhang SX, Watkins T, et al. Multicenter evaluation
8. Peralta G, Sanchez MB, Garrido JC, et al. Impact of antibiotic of Candida QuickFISH BC for identification of Candida species
resistance and of adequate empirical antibiotic treatment in the directly from blood culture bottles. J Clin Microbiol.
prognosis of patients with Escherichia coli bacteraemia. J 2015;53:1672–1676.
Antimicrob Chemother. 2007;60:855–863. 27. Ly T, Gulia J, Pyrgos V, et al. Impact upon clinical outcomes of
9. MacVane SH, Nolte FS. Benefits of adding a rapid PCR-based blood translation of PNA FISH-generated laboratory data from the clinical
culture identification panel to an established antimicrobial stew- microbiology bench to bedside in real time. Ther Clin Risk Manag.
ardship program. J Clin Microbiol. 2016;54:2455–2463. 2008;4:637–640.
10. Peters RPH, Savelkoul PHM, Simoons-Smit AM, et al. Faster identi- 28. Forrest GN, Mehta S, Weekes E, et al. Impact of rapid in situ
fication of pathogens in positive blood cultures by fluorescence in hybridization testing on coagulase-negative staphylococci positive
situ hybridization in routine practice. J Clin Microbiol. 2006;44:119– blood cultures. J Antimicrob Chemother. 2006;58:154–158.
123. 29. Stone NRH, Gorton RL, Barker K, et al. Evaluation of PNA-FISH yeast
11. Chapin K, Musgnug M. Evaluation of three rapid methods for the traffic light for rapid identification of yeast directly from positive
direct identification of Staphylococcus aureus from positive blood blood cultures and assessment of clinical impact. J Clin Microbiol.
cultures. J Clin Microbiol. 2003;41:4324–4327. 2013;51:1301–1302.
12. Salimnia H, Fairfax MR, Lephart PR, et al. Evaluation of the filmarray 30. Holtzman C, Whitney D, Barlam T, et al. Assessment of impact of
blood culture identification panel: results of a multicenter con- peptide nucleic acid fluorescence in situ hybridization for rapid
trolled trial. J Clin Microbiol. 2016;54:687–698. identification of coagulase-negative staphylococci in the absence
13. Arroyo MA, Denys GA. Parallel evaluation of the maldi Sepsityper of antimicrobial stewardship intervention. J Clin Microbiol.
and verigene BC-GN assays for rapid identification of gram-nega- 2011;49:1581–1582.
tive bacilli from positive blood cultures. J Clin Microbiol. 31. Frye AM, Baker CA, Rustvold DL, et al. Clinical impact of a real-time
2017;55:2708–2718. PCR assay for rapid identification of Staphylococcal bacteremia. J
14. Kothari A, Morgan M, Haake DA. Emerging technologies for rapid Clin Microbiol. 2012;50:127–133.
identification of bloodstream pathogens. Clin Infect Dis. 32. Maurer FP, Christner M, Hentschke M, et al. Advances in rapid
2014;59:272–278. identification and susceptibility testing of bacteria in the clinical
• This nice review helped us establish a feel for what had gone microbiology laboratory: implications for patient care and antimi-
before as allowing us to set a baseline for what we reviewed. crobial stewardship programs. Infect Dis Rep. 2017;9:18–27.
15. Ecker DJ, Sampath R, Li H, et al. New technology for rapid mole- 33. Accelerate Diagnostics. Morphokinetic Cellular Analysis - Accelerate
cular diagnosis of bloodstream infections. Expert Rev Mol Diagn. PhenoTM System. [cited 2018 May 22]. Available from: http://accel
2010;10:399–415. eratediagnostics.com/technology/mca/
16. Sinha M, Jupe J, Mack H, et al. Emerging technologies for molecular 34. Charnot-Katsikas A, Tesic V, Love N, et al. Use of Accelerate
diagnosis of sepsis. Clin Microbiol Rev. 2018;31:e00089–17. PhenoTM system for identification and antimicrobial susceptibility
17. Ramilo O, Allman W, Chung W, et al. Gene expression patterns in blood testing (ID/AST) of pathogens in positive blood cultures and impact
leukocytes discriminate patients with acute infections. Blood. on time to results and workflow. J Clin Microbiol. 2017.
2007;109:2066–2077. DOI:10.1128/JCM.01166-17
18. Pan Q, Luo F, Liu M, et al. Oligonucleotide aptamers: promising and 35. Marschal M, Bachmaier J, Autenrieth, et al. Evaluation of the
powerful diagnostic and therapeutic tools for infectious diseases. J Accelerate Pheno system for fast identification and antimicrobial
Infect. 2018. DOI:10.1016/j.jinf.2018.04.007 susceptibility testing from positive blood cultures in bloodstream
19. Mejias A, Ramilo O. Transcriptional profiling in infectious diseases: infections caused by gram-negative pathogens. J Clin Microbiol.
ready for prime time? J Infect. 2014;68:S94–S99. 2017;55:2116–2126.
20. Ashkenazi-Hoffnung L, Oved K, Navon R, et al. A host-protein 36. Pancholi P, Carroll KC, Buchan BW, et al. Multicenter evaluation of
signature is superior to other biomarkers for differentiating the Accelerate PhenoTestTM BC kit for rapid identification and
between bacterial and viral disease in patients with respiratory phenotypic antimicrobial susceptibility testing using morphoki-
infection and fever without source: a prospective observational netic cellular analysis. J Clin Microbiol. 2018;23. DOI:10.1128/
study. Eur J Clin Microbiol Infect Dis. 1007;10096–18. DOI:10.1007/ JCM.01329-17
s10096-018-3261-3 37. Pantel A, Monier J, Lavigne J-P. Performance of the Accelerate
21. Deck MK, Anderson ES, Buckner RJ, et al. Multicenter evaluation of PhenoTM system for identification and antimicrobial susceptibility
the Staphylococcus QuickFISH method for simultaneous identifica- testing of a panel of multidrug-resistant Gram-negative bacilli
tion of Staphylococcus aureus and coagulase-negative staphylo- directly from positive blood cultures. J Antimicrob Chemother.
cocci directly from blood culture bottles in less than 30 minutes. 2018;032–032.
J Clin Microbiol. 2012;50:1994–1998. 38. Afshari A, Schrenzel J, Ieven M, et al. Bench-to-bedside review:
22. Carretto E, Bardaro M, Russello G, et al. Comparison of the rapid molecular diagnostics for bloodstream infection - a new
Staphylococcus QuickFISH BC test with the tube coagulase test frontier? Crit Care. 2012;16:222.
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS 553

39. Case RJ, Boucher Y, Dahllof I, et al. Use of 16S rRNA and rpoB genes as staphylococcal cassette chromosome element lacking the mecA
molecular markers for microbial ecology studies. Appl Environ gene. J Clin Microbiol. 2011;49:722–724.
Microbiol. 2007;73:278–288. 59. Arbefeville SS, Zhang K, Kroegar JS, et al. Prevalence and genetic
40. Moore MS, McCarroll MG, McCann CD, et al. Direct screening of blood by relatedness of methicillin susceptible Staphylococcus aureus isolates
PCR and pyrosequencing for a 16s rDNA target from ED and ICU detected by the Xpert MRSA nasal assay. J Clin Microbiol.
patients being evaluated for bloodstream infection. J Clin Microbiol. 2011;49:2996–2999.
2016;54:99–105. 60. Cognitor minus. [cited 2018 May 22]. Available from: http://www.
41. Molzym SepiTest. [cited 2018 May 22]. Available from: http://www. momentumbio.co.uk/
sepsitest.com/ 61. Zweitzig DR, Riccardello NM, Sodowich BI, et al. Characterization of
42. Loonen AJM, de Jager CPC, Tosserams J, et al. Biomarkers and a novel DNA polymerase activity assay enabling sensitive, quanti-
molecular analysis to improve bloodstream infection diagnostics tative and universal detection of viable microbes. Nucleic Acids
in an emergency care unit. PLoS One. 2014;9:1–7. Res. 2012;40:1–12.
43. Warhurst G, Dunn G, Chadwick P, et al. Rapid detection of health- 62. Zweitzig DR, Riccardello NM, Morrison J, et al. Measurement of
care-associated bloodstream infection in critical care using multi- microbial DNA polymerase activity enables detection and growth
pathogen real-time polymerase chain reaction technology: a diag- monitoring of microbes from clinical blood cultures. PLoS One.
nostic accuracy study and systematic review. Health Technol Assess 2013;8:1–9.
(Rockv). 2015;19:1–141. 63. Zweitzig DR, Sodowich BI, Riccardello NM, et al. Feasibility of a
44. UK Public Health England. Standards for microbiology investiga- novel approach for rapid detection of simulated bloodstream infec-
tions (SMI) - commercial and in-house diagnostic tests: evaluations tions via enzymatic template generation and amplification (ETGA)-
and validations. Bacteriology. 2014;B 55:1–21. mediated measurement of microbial DNA polymerase activity. J
45. Phusion Blood Direct PCR Kit. [cited 2018 May 22]. Available from: Mol Diagn. 2013;15:319–330.
https://www.thermofisher.com/order/catalog/product/F547S 64. Dryden M, Sitjar A, Gunning Z, et al. Can rapid negative exclusion
46. Fuehrer HP, Fally MA, Habler VE, et al. Novel nested direct PCR of blood cultures by a molecular method, enzyme template gen-
technique for malaria diagnosis using filter paper samples. J Clin
Microbiol. 2011;49:1628–1630.
®
eration and amplification technique (Cognitor Minus), aid antimi-
crobial stewardship? Int J Pharm Pract. 2017.
®
47. r-biopharm RIDA GENE MRSA real-time PCR. [cited 2018 May 22]. • This paper describes a totally new approach to blood cultures
Available from: https://clinical.r-biopharm.com/products/ridagene- and how important knowing a guaranteed negative can
mrsa/ impact antimibrobial stewardship.
®
48. Xpert MRSA/SA BC. [cited 2018 May 22]. Available from: http:// 65. Frolov AE, Godwin AK, Favorova OO. Differential gene expression
www.cepheid.com/us/cepheid-solutions/clinical-ivd-tests/health analysis by DNA microarray technology and its application in
care-associated-infections/xpert-mrsa-sa-bc molecular oncology. Mol Biol. 2003;37:486–494.
49. Scanvic A, Courdavault L, Sollet J-P, et al. Intérêt de la PCR temps 66. Revel AT, Talaat AM, Norgard MV. DNA microarray analysis of
®
réel cepheid xpert SAMR/SA sur GeneXpert DX system dans la
prise en charge de la bactériémie à staphylocoque. Pathol Biol
differential gene expression in Borrelia burgdorferi, the Lyme dis-
ease spirochete. Proc Natl Acad Sci U S A. 2002;99:1562–1567.
(Paris). 2011;59:67–72. 67. Jordan JA, Jones-Laughner J, Durso MB. Utility of pyrosequencing
50. Spencer DH, Sellenriek P, Burnham CAD. Validation and implemen- in identifying bacteria directly from positive blood culture bottles. J
tation of the GeneXpert MRSA/SA blood culture assay in a pediatric Clin Microbiol. 2009;47:368–372.
setting. Am J Clin Pathol. 2011;136:690–694. 68. Liesenfeld O, Lehman L, Hunfeld K-P, et al. Molecular diagnosis of
51. Davies J, Gordon CL, Tong SYC, et al. Impact of results of a rapid sepsis: new aspects and recent developments. Eur J Microbiol
Staphylococcus aureus diagnostic test on prescribing of antibiotics Immunol. 2014;4:1–25.
for patients with clustered Gram-positive cocci in blood cultures. J 69. Decuypere S, Meehan CJ, van puyvelde S, et al. Diagnosis of
Clin Microbiol. 2012;50:2056–2058. bacterial bloodstream infections: a 16s metagenomics approach.
52. Buchan BW, Ginocchio CC, Manii R, et al. Multiplex identification of PLoS Negl Trop Dis. 2016;10:1–12.
Gram-positive bacteria and resistance determinants directly from 70. Deurenberg RH, Bathoorn E, Chlebowicz MA, et al. Application of
positive blood culture broths: evaluation of an automated micro- next generation sequencing in clinical microbiology and infection
array-based nucleic acid test. PLoS Med. 2013;10. prevention. J Biotechnol. 2017;243:16–24.
53. BD max MRSA/SA. [cited 2018 May 22]. Available from: https:// 71. Nanosphere verigene. [cited 2018 May 22]. Available from: https://
www.bd.com/en-uk/products/molecular-diagnostics/molecular-sys www.luminexcorp.com/eu/clinical/our-technology/verigene-
tems/max-system/max-mrsa-xt-with-extended-detection- technology/
technology 72. Beal SG, Ciurca J, Smith G, et al. Evaluation of the Nanosphere
54. Lee J, Park YJ, Park DJ, et al. Evaluation of bd max staph sr assay Verigene Gram-positive blood culture assay with the Versatrek
for differentiating between staphylococcus aureus and coagu- blood culture system and assessment of possible impact on
lase-negative staphylococci and determining methicillin resis- selected patients. J Clin Microbiol. 2013;51:3988–3992.
tance directly from positive blood cultures. Ann Lab Med. 73. Mestas J, Polanco CM, Felsenstein S, et al. Performance of the
2017;37:39–44. Verigene Gram-positive blood culture assay for direct detection of
55. Huletsky A, Giroux R, Rossbach V, et al. New real-time PCR assay Gram-positive organisms and resistance markers in a pediatric
for rapid detection of methicillin resistant Staphylococcus aureus hospital. J Clin Microbiol. 2014;52:283–287.
directly from specimens containing a mixture of staphylococci. 74. Kikuchi K, Matsuda M, Iguchi S, et al. Potential impact of rapid blood
Society. 2004;42:1875–1884. culture testing for Gram-positive bacteremia in Japan with the Verigene
56. Dalpke AH, Hofko M, Hamilton F, et al. Evaluation of the BD max Gram-positive blood culture test. Can J Infect Dis Med Microbiol. 2017.
StaphSR assay for rapid identification of Staphylococcus aureus and 75. Hill JT, Tran K-DT, Barton KL, et al. Evaluation of the Nanosphere
methicillin resistant S. aureus in positive blood culture broths. J Clin Verigene bc-gn assay for direct identification of Gram-negative
Microbiol. 2015;53:3630–3632. bacilli and antibiotic resistance markers from positive blood cul-
57. Petersen A, Stegger M, Heltberg O, et al. Epidemiology of methi- tures and potential impact for more-rapid antibiotic interventions.
cillin resistant Staphylococcus aureus carrying the novel mecC gene J Clin Microbiol. 2014;52:3805–3807.
in Denmark corroborates a zoonotic reservoir with transmission to 76. Ledeboer NA, Lopansri BK, Dhiman N, et al. Identification of Gram-
humans. Clin Microbiol Infect. 2013;19:E16–E22. negative bacteria and genetic resistance determinants from positive
58. Blanc DS, Basset P, Nahimana-Tessemo I, et al. High proportion of blood culture broths by use of the Verigene Gram-negative blood
wrongly identified methicillin resistant Staphylococcus aureus car- culture multiplex microarray-based molecular assay. J Clin Microbiol.
riers by use of a rapid commercial PCR assay due to presence of 2015;53:2460–2472.
554 S. POOLE ET AL.

77. Walker T, Dumadag S, Lee CJ, et al. Clinical impact after laboratory the routine practice of clinical microbiology. Clin Microbiol Rev.
implementation of the Verigene Gram-negative bacteria microarray 2013;26:547–603.
for positive blood cultures. J Clin Microbiol. 2016;54:1789–1796. 98. Yapo-Kouadio C, Jeddi F, Cassagne C, et al. Performance assess-
78. Sothoron C, Ferreira J, Guzman N, et al. A stewardship approach to ment of two lysis methods for direct identification of yeasts from
optimize antimicrobial therapy through use of a rapid microarray clinical blood cultures using MALDI-TOF mass spectrometry. Med
assay on blood cultures positive for Gram-negative bacteria. J Clin Mycol 2018;185–192.
Microbiol. 2015;53:3627–3629. 99. Yonetani S, Ohnishi H, Ohkusu K, et al. Direct identification of
79. Beal SG, Thomas C, Dhiman N, et al. Antibiotic utilization improve- microorganisms from positive blood cultures by MALDI-TOF MS
ment with the Nanosphere Verigene Gram-positive blood culture using an in-house saponin method. Int J Infect Dis. 2016;52:37–
assay. Proc (Bayl Univ Med Cent). 2015;28:139–143. 42.
80. Biofire filmarray DX BCID. [cited 2018 May 22]. Available from: 100. Idelevich EA, Grunastel SB, Wullenweber J, et al. Rapid identifi-
http://www.biofiredx.com/products/the-filmarray-panels/#blood- cation of microorganisms from positive blood cultures by
culture MALDI-TOF mass spectrometry subsequent to very short-term
81. Blaschke AJ, Heyrend C, Byington CL, et al. Rapid identification of incubation on solid medium. Clin Microbiol Infect.
pathogens from positive blood cultures by multiplex PCR using the 2014;20:1001–1006.
Filmarray system. Diagn Microbiol Infect Dis. 2013;74:349–355. 101. Curtoni A, Cipriani R, Marra ES, et al. Rapid Identification of micro-
82. Altun O, Almuhayawi M, Ullberg M, et al. Clinical evaluation of the organisms from positive blood culture by MALDI-TOF MS after
Filmarray blood culture identification panel in identification of short-term incubation on solid medium. Curr Microbiol.
bacteria and yeasts from positive blood culture bottles. J Clin 2017;74:97–102.
Microbiol. 2013;51:4130–4136. 102. Kohlmann R, Hoffmann A, Geis G, et al. MALDI-TOF mass spectro-
83. Zheng X, Polanco W, Carter D. Rapid identification of pathogens metry following short incubation on a solid medium is a valuable
from pediatric blood cultures by use of the Filmarray blood culture tool for rapid pathogen identification from positive blood cultures.
identification panel. J Clin Microbiol. 2014;52:4368–4371. Int J Med Microbiol. 2015;305:469–479.
84. Banerjee R, Teng CB, Cunningham SA, et al. Randomized trial of 103. Konnerth S, Rademacher G, Suerbaum S, et al. Identification of
rapid multiplex polymerase chain reaction-based blood culture pathogens from blood culture bottles in spiked and clinical sam-
identification and susceptibility testing. Clin Infect Dis. ples using matrix-assisted laser desorption ionization time-of-flight
2015;61:1071–1080. mass-spectrometry analysis. BMC Res Notes. 2014;7:1–4.
85. Paolucci M, Foschi C, Tamburini MV, et al. Comparison between 104. Fiori B, D'Inzeo T, Di Florio V, et al. Performance of two resin-
MALDI-TOF MS and Filmarray blood culture identification panel for containing blood culture media in detection of bloodstream infec-
rapid identification of yeast from positive blood culture. J Microbiol tions and in direct matrix-assisted laser desorption ionization-time
Methods. 2014;104:92–93. of flight mass spectrometry (MALDI-TOF MS) broth assays for iso-
86. Blaschke AJ, Heyrend C, Byington CL, et al. Rapid identification of late identification: clinical comp. J Clin Microbiol. 2014;52:3558–
pathogens from positive blood cultures by multiplex polymerase 3567.
chain reaction using the Filmarray system. Diagn Microbiol Infect 105. Tadros M, Petrich A. Evaluation of MALDI-TOF mass spectrometry
Dis. 2018;74:349–355. and Sepsityper kitTM for the direct identification of organisms from
87. GenMark’s ePlex blood culture identification (BCID). [cited 2018 sterile body fluids in a Canadian pediatric hospital. Can J Infect Dis
May 22]. Available from: https://www.genmarkdx.com/int/solu Med Microbiol. 2013;24:191–194.
tions/panels/eplex-panels/blood-culture-identification-panels 106. Bruker MBT Sepsityper IVD-CE kit. [cited 2018 May 22]. Available
88. Maubon D, Dard C, Garnaud C, et al. Profile of genmark’s ePlex ® from: https://www.bruker.com/products/mass-spectrometry-and-
blood culture identification fungal pathogen panel. Expert Rev Mol separations/ivd-ce-certified-maldi-biotyper/mbt-sepsityper-ivd-kit.
Diagn. 2018;18:119–132. html
89. Curetis Unyvero BCU. [cited 2018 May 22]. Available from: http:// 107. Morgenthaler NG, Kostrzewa M. Rapid identification of pathogens
www.unyvero.com/en/applications/bcu-application.html in positive blood culture of patients with sepsis: review and meta-
90. Sepsis flow chip assay. [cited 2018 May 22]. Available from: http:// analysis of the performance of the Sepsityper kit. Int J Microbiol.
www.masterdiagnostica.com/en-gb/products/moleculardiagnos 2015;2015.
tickits/dnaflowtechnology/sepsisflowchip.aspx 108. Faron ML, Buchan BW, Ledeboer NA. Matrix-assisted desorption
91. Galiana, A., Coy J, Gimeno A, et al. Evaluation of the sepsis flow ionization time of flight mass spectrometry for the use with posi-
chip assay for the diagnosis of blood infections. PLoS One. 2017;1– tive blood cultures: methodology, performance, and optimization. J
12. Clin Microbiol. 2017. DOI:10.1128/JCM.00868-17
92. Belkum AV, Victoria. G, Maud. A, et al. MALDI TOF MS for Microbial 109. DeMarco ML, Ford BA. Beyond identification: emerging and future
Identification in Clinical Microbiology. In: Persing DH, Tenover FC, uses for MALDI-TOF mass spectrometry in the clinical microbiology
Hayden R, ed.. Molecular Microbiology - Diagnostic Principles and laboratory. Clin Lab Med. 2013;33:611–628.
Practice. ASM Press; 2016. p. 92–101. 110. Vella A, De Carolis E, Vaccaro L, et al. Rapid antifungal susceptibility
93. Buchan BW, Riebe KM, Ledeboer NA. Comparison of the MALDI testing by matrix-assisted laser desorption ionization-time of flight
biotyper system using Sepsityper specimen processing to routine mass spectrometry analysis. J Clin Microbiol. 2013;51:2964–2969.
microbiological methods for identification of bacteria from positive 111. Lee AWT, Lam JKS, Lam RKW, et al. Comprehensive evaluation of
blood culture bottles. J Clin Microbiol. 2012;50:346–352. the MBT STAR-BL module for simultaneous bacterial identification
94. Croxatto A, Prod’hom G, Greub G. Applications of MALDI-TOF mass and β-lactamase-mediated resistance detection in Gram-negative
spectrometry in clinical diagnostic microbiology. FEMS Microbiol rods from cultured isolates and positive blood cultures. Front
Rev. 2012;36:380–407. Microbiol. 2018;9.
95. Hariu M, Watanabe Y, Oikawa N, et al. Usefulness of matrix-assisted 112. Papagiannitsis CC, Studentova V, Izdebski R, et al. Matrix-assisted
laser desorption ionization time-of-flight mass spectrometry to laser desorption ionization-time of flight mass spectrometry mer-
identify pathogens, including polymicrobial samples, directly from openem hydrolysis assay with NH4HCO3, a reliable tool for direct
blood culture broths. Infection and Drug Resistance. 2017;115–120. detection of carbapenemase activity. J Clin Microbiol.
96. Vincent JL, Brealey D, Libert N, et al. Rapid diagnosis of infection in 2015;53:1731–1735.
the critically ill, a multicenter study of molecular detection in 113. Sparbier K, Lange C, Jung J, et al. Maldi biotyper-based rapid
bloodstream infections, pneumonia, and sterile site infections. resistance detection by stable-isotope labeling. J Clin Microbiol.
Crit Care Med. 2015;43:2283–2291. 2013;51:3741–3748.
97. Clark AE, Kaleta EJ, Arora A, et al. Matrix-assisted laser desorption 114. Hrabák J, Chudác Ǩ ová E, Walková R. Matrix-assisted laser deso-
ionization-time of flight mass spectrometry: a fundamental shift in rption ionization-time of flight (MALDITOF) mass spectrometry for
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS 555

detection of antibiotic resistance mechanisms: from research to spectrometry (SIFT-MS) and comparison with the bact/ALERT auto-
routine diagnosis. Clin Microbiol Rev. 2013;26:103–114. mated blood culture system. J Microbiol Methods. 2006;65:628–631.
115. Jung JS, Hamacher C, Gross B, et al. Evaluation of a semi-quantita- 139. Dolch ME, Hornuss C, Klocke C, et al. Volatile compound profiling
tive MALDI-TOF MS method for rapid antimicrobial susceptibility for the identification of Gram-negative bacteria by ion-molecule
testing of positive blood cultures. J Clin Microbiol. 2016;54: reaction-mass spectrometry. J Appl Microbiol. 2012;113:1097–1105.
JCM.01131–16. 140. Wiesner K, Jaremek M, Pohle R, et al. Monitoring of bacterial
116. Beutz M, Sherman G, Mayfield J, et al. Clinical utility of blood growth and rapid evaluation of antibiotic susceptibility by head-
cultures drawn from central vein catheters and peripheral veni- space gas analysis. Procedia Eng. 2014;87:332–335.
puncture in critically ill medical patients. Chest. 2003;123:854–861. 141. Dolch ME, Janitza S, Boulesteix AL, et al. Gram-negative and -posi-
117. ®
Roche LightCycler SeptiFast. [cited 2018 May 22]. Available from:
https://molecular.roche.com/assays/lightcycler-septifast-test-
tive bacteria differentiation in blood culture samples by headspace
volatile compound analysis. J Biol Res. 2016;23:3.
mgrade/ 142. Lim SH, Mix S, Xu Z, et al. Colorimetric sensor array allows fast
118. Korber F, Zeller I, Grunstaudl M, et al. SeptiFast versus blood detection and simultaneous identification of sepsis-causing bac-
culture in clinical routine – A report on 3 years experience. Wien teria in spiked blood culture. J Clin Microbiol. 2014;52:592–598.
Klin Wochenschr. 2017;129:427–434. 143. Shrestha NK, Lim SH, Wilson DA, et al. The combined rapid detec-
119. Ziegler I, Josefson P, Olcén P, et al. Quantitative data from the tion and species level identification of yeasts in simulated blood
SeptiFast real-time PCR is associated with disease severity in culture using a colorimetric sensor array. PLoS One. 2017;12:1–12.
patients with sepsis. BMC Infect Dis. 2014;14:155. 144. Q-linea astrid. [cited 2018 May 22]. Available from: https://qlinea.
120. Mongelli G, Romeo MA, Denaro C, et al. Added value of multi- com/
pathogen probe-based real-time PCR SeptiFast in the rapid diag- 145. Hardenbol P, Baner J, Jain M, et al. Multiplexed genotyping with
nosis of bloodstream infections in patients with bacteraemia. J Med sequence-tagged molecular inversion probes. Nat Biotechnol.
Microbiol. 2015;64:670–675. 2003;21:673–678.
121. Pasqualini L, Mencacci A, Leli C, et al. Diagnostic performance of a 146. Nilsson M, Malmgren H, Samiotaki M, et al. Padlock probes: circu-
multiple real-time PCR assay in patients with suspected sepsis larizing oligonucleotides for localized DNA detection. Science.
hospitalized in an internal medicine ward. J Clin Microbiol. 1994;265:2085–2088.
2012;50:1285–1288. 147. Baner J, Nilsson M, Mendel-Hartvig M, et al. Signal amplification of
122. Huttunen R, Syrjänen J, Vuento R, et al. Current concepts in the padlock probes by rolling circle replication. Nucleic Acids Res.
diagnosis of blood stream infections: are novel molecular methods 1998;26:5073–5078.
useful in clinical practice? Int J Infect Dis. 2013;17:934–938. 148. Dahl F, Baner J, Gullberg M, et al. Circle-to-circle amplification for
123. SeeGene magicplex. [cited 2018 May 22]. Available from: http:// precise and sensitive DNA analysis. Proc Natl Acad Sci U S A.
www.seegene.com 2004;101:4548–4553.
124. Marco D, Scolfaro C, Colombo S, et al MagicplexTM sepsis real-time 149. Morck J,Goransson C, Goransson J, et al. Evaluation of a new plat-
test to improve bloodstream infection diagnostics in children. Eur J form for rapid identification and susceptibility testing of sepsis
Pediatr. 2016;175:1107–1111. pathogens. Poster #425 at ASM. 2017.
125. T2 biosystems. [cited 2018 May 22]. Available from: https://www. 150. Qvella Fast Id. [cited 2018 May 22]. Available from: https://www.
t2biosystems.com qvella.com/why-us
126. Tsalik EL, Jones D, Nicholson B, et al. Multiplex PCR to diagnose 151. Culbreath K, Petti C. Balancing enthusiasm for innovative technol-
bloodstream infections in patients admitted from the emergency ogies with optimizing value: an approach to adopt new laboratory
department with sepsis. J Clin Microbiol. 2010;48:26–33. tests for infectious diseases using bloodstream infections as exem-
127. Zervou FN, Zacharioudakis IM, Kurpewski J, et al. T2 magnetic reso- plar. Ofid. 2015;2:1–8.
nance for fungal diagnosis. Methods Mol Biol. 2017;1508:305–319. •• This paper explains a good method to work through in the
128. Hamula CL, Hughes K, Fisher BT, et al. T2Candida provides rapid adoption of a new diagnostic technology ensuring all teams
and accurate species identification in pediatric cases of candide- are involved when assessing the value of its potential
mia. Am J Clin Pathol. 2016;145:858–861. implementation.
129. Perlin DS, Wiederhold NP. Culture-independent molecular methods 152. Public Health England. UK standards for microbiology investi-
for detection of antifungal resistance mechanisms and fungal iden- gations: investigation of blood cultures (for organisms other
tification. J Infect Dis. 2017;216:S458–S465. than Mycobacterium species). UK Stand Microbiol Investig.
130. Clancy CJ, Nguyen MH. Diagnosing invasive candidiasis. J Clin 2014;B 37.
Microbiol. 2018. DOI:10.1128/JCM.01909-17 153. Venturelli C, Righi E, Borsari L, et al. Impact of pre-analytical time on
131. Clancy CJ, Pappas PG, Vazquez J, et al. Detecting infections rapidly the recovery of pathogens from blood cultures: results from a large
and easily for candidemia trial, part 2 (direct2): a prospective, multi- retrospective survey. PLoS One. 2017;1–11.
center study of the T2candida panel. Clin Infect Dis. 2018;1095–1095. 154. Kerremans JJ, Van Der Bij AK, Goessens W, et al. Immediate incuba-
132. Templier V, Livache T, boisset S, et al. Biochips for direct detection tion of blood cultures outside routine laboratory hours of opera-
and identification of bacteria in blood culture-like conditions. tion accelerates antibiotic switching. J Clin Microbiol.
Nature Scientific reports. 2017;1–10. 2009;47:3520–3523.
133. Wilson MR, Chiu CY. Actionable diagnosis of neuroleptospirosis by 155. Weinbren M, Heathcote R, Collins M. National blood culture standards
next-generation sequencing. N Engl J Med. 2014;370:2408–2417. and the multidisciplinary approach. Biomed Sci. 2016;194–198.
134. Wilson MR, DeRisi JL. Chronic meningitis investigated via metage- •• The authors describe a pan-pathology and multidisciplinary
nomic next-generation sequencing. JAMA Neurol. 2018;E1-E9. approach to blood culture that has much merit.
135. Quick J, Loman NJ, Duraffour S, et al. Real-time, portable genome 156. Van Der Velden LB, Vos FJ, Mouton JW, et al. Clinical impact of
sequencing for ebola surveillance. Nature. 2016;530:228–232. preincubation of blood cultures at 37°C. J Clin Microbiol.
136. SpecID. [cited 2018 May 22]. Available from: https://www.speci 2011;49:275–280.
ficdx.com/ 157. Cohen JF, Korevaar DA, Altman DG, et al. STARD 2015 guidelines for
137. Rees C, Burklund A, Stefanuto P-H, et al. Comprehensive volatile reporting diagnostic accuracy studies: explanation and elaboration.
metabolic fingerprinting of bacterial and fungal pathogen groups. BMJ Open. 2016;6(1–18).
J Breath Res. 2017. • This paper highlights the need for good quality analytical
138. Scotter JM, Allardyce RA, Langford VS, et al. The rapid evaluation of evaluations of new diagnostic technologies and proposed
bacterial growth in blood cultures by selected ion flow tube–mass standardisation.

You might also like