Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Papers in Press. Published October 23, 2018 as doi:10.1373/clinchem.2016.

266536
The latest version is at http://hwmaint.clinchem.aaccjnls.org/cgi/doi/10.1373/clinchem.2016.266536

Clinical Chemistry 65:3


000 – 000 (2019) Reviews

Next-Generation Sequencing for Biodefense:


Biothreat Detection, Forensics, and the Clinic
Timothy D. Minogue,1*† Jeffrey W. Koehler,1† Christopher P. Stefan,1† and Turner A. Conrad1

BACKGROUND: Next-generation sequencing (NGS) is lus anthracis spores to various media outlets and political
revolutionizing a variety of molecular biology fields in- figures, causing infections in 22 individuals and 5 deaths
cluding bioforensics, biosurveillance, and infectious dis- (1 ). This event resulted in (a) a 7-year investigation and
ease diagnostics. For pathogen detection, the ability to the treatment of over 10000 people with antibiotic pro-
sequence all nucleic acids in a sample allows near limitless phylaxis, (b) the collection and testing of 120000 clinical
multiplexability, free from a priori knowledge regarding and environmental samples, and (c) the quarantine of 1.8
an etiologic agent as is typically required for targeted million mailed items. These attacks resulted in billions of
molecular assays such as real-time PCR. Furthermore, research dollars being allocated toward extant and nas-
sequencing capabilities can generate in depth genomic cent biodefense efforts (2 ), including efforts to enhance
information, allowing detailed molecular epidemiologi- bioforensics to support criminal prosecutions. Termed
cal studies and bioforensics analysis, which is critical for the Amerithrax case, this investigation was instrumental
source agent identification in a biothreat outbreak. How- in demonstrating capabilities and readiness in the fields
ever, lack of analytical specificity, inherent to NGS, pres- of biosurveillance, clinical diagnostics, and bioforen-
ents challenges for regulated applications such as clinical sics in response to a bioterrorism event. While next-
diagnostics and molecular attribution. generation sequencing (NGS)2 was not used directly
in this case, the lineage of NGS applications in the
CONTENT: Here, we discuss NGS applications in the con- biothreat surveillance and diagnostics context are a
text of preparedness and biothreat readiness. Specifically, we direct result of increased research funding stemming
investigate current and future applications of NGS technol- from this incident.
ogies to affect the fields of biosurveillance, bioforensics, and Staple assays for rapid pathogen detection at the on-
clinical diagnostics with specific focus on biodefense. set of a biothreat public health crisis, such as the Ameri-
thrax case, can include PCR assays (3, 4 ), enzyme-linked
SUMMARY: Overall, there are many advantages to the im- immunosorbent assays (5 ), and lateral flow immunoas-
plementation of NGS for preparedness and readiness says (6 ). While these approaches are excellent for patho-
against biowarfare agents, from forensics to diagnostics. gen identification and provide clinically actionable data,
However, appropriate caveats must be associated with these techniques have limited multiplexability and re-
any technology. This includes NGS. While NGS is not quire a priori knowledge of the causative agent. These
the panacea replacing all molecular techniques, it will methods can also leave an information gap concerning
greatly enhance the ability to detect, characterize, and the genetic makeup of an organism, a critical piece to
diagnose biowarfare agents, thus providing an excellent bioforensics solutions. Sequencing specific genes or
addition to the biodefense toolbox of biosurveillance, genomic regions can bridge this forensics gap, but, before
bioforensics, and biothreat diagnosis. the advent of NGS technology, prior detection assays
© 2018 American Association for Clinical Chemistry failed to provide the needed capabilities of producing
high levels of quality genetic information rapidly.
Historically, modern sequencing began in 1977
with the development of Sanger sequencing (7, 8 ) (Fig.
Following the terrorist attacks of September 11, 2001, an
1). Since the introduction of sequencing by synthesis,
unidentified individual sent letters containing live Bacil-
advances in the technology have outpaced Moore’s law.
Second-generation technologies followed Sanger in the
mid-2000s, with pyrosequencing, sequencing by liga-
1
Diagnostic Systems Division, United States Army Medical Research Institute of Infectious tion, and reversible terminator chemistry sequencing.
Diseases, Fort Detrick, MD. This technology development allowed efficient sequenc-
* Address correspondence to this author at: United States Army Medical Research Institute
of Infectious Diseases, Diagnostic Systems Division, 1425 Porter Street, Fort Detrick, MD
21702. Fax 301-619-2492; e-mail timothy.d.minogue.civ@mail.mil.

These authors contributed equally to the writing of this review.
2
Received May 2, 2018; accepted June 22, 2018. Nonstandard abbreviations: NGS, next-generation sequencing; EVD, Ebola virus dis-
Previously published online at DOI: 10.1373/clinchem.2016.266536 ease; GMOs, genetically modified organisms; LDT, laboratory-developed test; FDA, Food
© 2018 American Association for Clinical Chemistry and Drug Administration.

Copyright (C) 2018 by The American Association for Clinical Chemistry


Reviews

Fig. 1. Advancements in sequencing technologies.


Considered the first generation of sequencers (blue), Sanger sequencing in 1977 provided the foundation for all sequencing applications.
New innovations with clonal amplification, parallel sequencing, and sequence by synthesis resulted in the second-generation technologies
(green). The introduction of long-read technologies led to the advent of third-generation sequencers (purple) with PacBio and Nanopore.

ing of all the nucleic acids within a sample, revolutioniz- NGS for Biosurveillance
ing how researchers approached biothreat and infectious
disease detection. Subsequent technology maturation re- Biosurveillance is a critical component of emerging infec-
sulted in the current wave of third-generation sequencers tious diseases and biothreat preparedness. Without cur-
focusing on single-molecule nucleic acid detection in real rent, real-time knowledge of the endemic circulating
time (Fig. 1). This approach obviates clonal amplifica- pathogens, it is impossible to prepare and/or appropri-
tion requirements common to previous generations of ately respond to an outbreak or biological event. In this
sequencers, reducing both amplification errors and time context, biosurveillance is defined as gathering, amal-
to answer (9 ). As technology continues to evolve, NGS is gamating, and interpreting data to communicate and es-
beginning to find a role in regulated clinical laboratories tablish actionable results for emerging and biothreat dis-
as an in vitro diagnostic. Overall, wide adoption and eases. Pathogen-agnostic NGS fits into this current
application of this technology is transitioning from basic paradigm for biosurveillance, augmenting and, in some
research applications to more regulated applications such cases replacing, traditional targeted assays such as real-
as biosurveillance, bioforensics, and clinical diagnostics time PCR and immunoassays to increase the detection
(Fig. 2). capability for unexpected or novel organisms.

2 Clinical Chemistry 65:3 (2019)


Biothreat Applications of Next-Generation Sequencing
Reviews

At the inception of the 2013–2015 outbreak, re-


searchers urgently sought to characterize the emerging
Ebola virus variant to better understand the virus’
genomic plasticity with regard to fielded diagnostic assays
and countermeasures. NGS-based sequencing early in
the outbreak identified a higher degree of diversity in this
isolate than expected, suggesting a negative effect on di-
agnostic assay performance (12 ). A larger characteriza-
tion of circulating Ebola virus isolates showed this plas-
ticity was not as significant as earlier believed (13 ). This
information coupled with empiric evaluations of existing
diagnostics (14 ) found the effect on fielded Ebola virus
assays was minimal. However, the initial genomic char-
acterizations raised critical concerns that, when ad-
dressed, resulted in higher confidence in assay results.
As the outbreak progressed, NGS capabilities be-
Fig. 2. Application of NGS for biothreat agents. came available in EVD-affected countries. Implementa-
The 3 main biothreat NGS applications discussed here include tion of NGS in near real time allowed for a better under-
biosurveillance (blue), bioforensics (green), and clinical use or standing of the Ebola virus transmission dynamics and
diagnostics (purple). For each use case, the general definition is aided in controlling the outbreak. For example, Christie
provided with the projected outputs for any NGS biothreat assays et al. (15 ) and Mate et al. (16 ) documented a suspected
highlighted. Biosurveillance principally deals with environmen- case of Ebola virus sexual transmission from an appar-
tal biothreat detection and surveillance with the mitigation and ently healthy survivor to his female partner. NGS and
tracking of an outbreak in as close to real time as possible. Biofo- genomic analysis confirmed the epidemiological hypoth-
rensics is more focused on meeting evidentiary standards to pro- esis of sexual transmission. These findings resulted in
vide attributional information. Biothreat clinical NGS diagnostics changes to CDC recommendations related to sexual be-
is centered on generating actionable information to affect patient havior of survivors to help end Ebola virus transmission
treatment. chains. Postoutbreak NGS sequencing of large infected
cohorts allowed detailed molecular epidemiological
tracking of the different transmission chains and helped
The discovery of a new rhabdovirus, Bas Congo vi- to understand the stability of the Ebola virus genome
rus, from an acute hemorrhagic fever patient in the Dem- during a sustained, large-scale outbreak (17–20 ). In Ni-
ocratic Republic of Congo, exemplifies this tiered-assay geria, for example, genomic sequencing and detailed con-
approach (10 ). Initial testing by real-time PCR for tact tracing from 20 EVD patients confirmed the Nige-
known hemorrhagic fever viruses was negative, necessi- rian outbreak resulted from a single introduction rather
tating NGS for pathogen identification. Deep sequenc- than multiple introduction events (18 ).
ing of the patient sample identified Bas Congo virus, a One important concern in the biosurveillance arena
previously unknown virus, generating a near complete is the potential for accidental or intentional release of
genome that formed the basis of a specific real-time RT- genetically modified biothreats designed for enhanced
PCR diagnostic assay and a pseudotyped virus for sero- pathogenicity, spread, or evasion of fielded countermea-
logical studies (10 ). In a similar case from 2008, Briese et sures (i.e., diagnostics, therapeutics, and vaccines). Tech-
al. used NGS to identify a novel arenavirus, Lujo virus, nology is advancing at a dynamic rate, so much so that
from a cluster of acute hemorrhagic fever patients in researchers have been able to generate an infectious
South Africa following transport of the index case from horsepox virus from synthetic DNA (21 ); this ability
the Zambia (11 ). raises the risk of someone using a similar strategy to re-
constitute other poxviruses. Such methods could be com-
NGS Biosurveillance for Biothreats bined with further genetic modifications to increase
pathogenicity, as others demonstrated for myxoma virus
Perhaps the best illustration of NGS biothreat biosurveil- by adding interluken 4 to modulate the host immune
lance is the most recent Ebola virus disease (EVD) out- response (22 ). While these technological advances gen-
break in West Africa, where researchers applied NGS in 2 erally improve public health, this benefit comes with an
ways: (a) characterizing the genome of the emerging increased risk of nefarious use. In cases in which these
pathogen, and (b) determining the transmission dynam- manipulations would be unexpected, NGS coupled with
ics of the virus. appropriate genome assembly could identify modifica-

Clinical Chemistry 65:3 (2019) 3


Reviews

tions that would be important to guide countermeasure a genome surveillance system to Guinea during the West
use and to begin assigning molecular attribution. Africa Ebola virus outbreak (29 ). This system was highly
There are no known instances of genetically modi- portable and could be transported by commercial airline
fied biothreat agents found in circulation; however, to Guinea, where it generated data within 2 days of ar-
the identification of genetically modified organisms rival. With use of PCR for genome enrichment before
(GMOs) in the agricultural field provides a framework sequencing, Ebola virus genomic information was avail-
for how NGS could be implemented for biothreats. The able in ⬍24 h of sample receipt (29 ). This rapid time to
European Union has strict GMO regulations and testing answer could affect fielded diagnostic assays during the
mechanisms in place, such as real-time PCR detection of outbreak, providing real-time analysis of sequence vari-
GMO-specific signatures (23, 24 ), to determine unau- ance affecting diagnostic assay efficacy.
thorized GMOs and to ensure compliance with regula- While resolving these technological challenges is an
tions. Rapid development of different GMOs now re- ongoing process, surveillance is the first important step in
quires a large number of assays to ensure regulatory a preparedness and readiness paradigm. Specifically, in
compliance. NGS, using its broad sequencing capability, the context of a biological incident, there is a defined
is beginning to fill this role (25–27 ). In this context, need to determine attribution of the source of the insult-
Paracchini et al. used NGS to characterize an unauthor- ing agent.
ized, genetically modified B. subtilis found in feed addi-
tives imported into Germany from China. Their analyses NGS Bioforensics
identified multiple genetic modifications and the inclu-
sion of 4 recombinant plasmids used to increase ribofla- Forensic microbiology is a relatively new scientific disci-
vin production (25 ). Although agricultural GMOs do pline created, for attributional purposes, to analyze evi-
not induce the same level of angst as an Ebola virus out- dence of bioterrorism, biocrime, or the inadvertent
break, there is a defined effect on economies and animal release of an etiologic agent. In bioforensics, agent iden-
welfare. tification is the starting point of an investigation. All
subsequent efforts seek to link organism-specific genetic
Challenges for NGS Biosurveillance information to a particular isolate, bacterial strain, or
even an individual person with sufficient confidence for
A major challenge for bringing NGS to the point of care legal review. The inception of modern bioforensic meth-
is the fielding of sequencing instruments in austere envi- ods began with Alec Jeffery’s development of human
ronments where most natural biothreats occur. In the case DNA fingerprinting, precipitating the current attribu-
of the West Africa EVD outbreak, there were substantial tional use of DNA for forensic samples (30 –32 ). Restric-
hurdles for NGS implementation including (a) instrument tion fragment length polymorphism and Sanger sequenc-
shipping, (b) setup and validation of functionality, (c) gen- ing of short tandem repeats are recent advances in this
eral maintenance, and (d) an uninterrupted power supply to field (33 ), but, similar to biosurveillance, resolving com-
complete the sequencing run. Furthermore, preparing se- plex DNA mixtures or identifying genetic variations can
quenceable libraries that employ enrichment or host back- be challenging with these methods (34 ). Some examples
ground reduction steps is highly technical, thus reducing of modern biocrimes, in which bioforensic experts used
assay utility in resource-limited, austere environments. the microorganism’s genetic material for attribution, in-
While NGS implementation challenges exist, these hurdles clude Salmonella typhimurium poisoning of salad bars
are not insurmountable. (35 ) and attempted murder with HIV-infected blood
Advances in NGS technologies, sequencing capac- (36 ) (Fig. 3). Resolution of these cases demonstrated the
ity, and analysis methods continue to expand the avail- merit of microbial bioforensics as a scientific discipline.
ability of this technology. For instance, the portability However, it was not until the 2001 intentional release of
and low cost of the MinION nanopore sequencer offers a B. anthracis spores in the Amerithrax letters that the value
potential solution to several of the resource limitations of biodefense microbial forensics came to fruition.
for biosurveillance applications. Greninger et al. demon- NGS found minimal use in the Amerithrax case pri-
strated a proof-of-concept study to identify Chikungu- marily due to the nascent nature of the technology; how-
nya virus, Ebola virus, and hepatitis C virus from human ever, it is worth discussing the application of Sanger se-
clinical samples by using the MinION and a web-based quencing in this case because this provides an excellent
analysis program (28 ). In this case a high-titer sample example of how NGS would be implemented currently. In
(107 copies/mL) from an Ebola virus–infected patient the Amerithrax investigational timeline, multiple locus
and a low titer (105 copies/mL) hepatitis C virus sample variable-number tandem repeat analysis subtyped the B. an-
had the first virus-specific reads identified within 10 and thracis used to the Ames strain (37 ). Subsequent Sanger-
40 min of sequencing, respectively (28 ). Another excel- based whole-genome sequencing narrowed the isolate on
lent example is from Quick et al., who built and deployed the basis of the presence of single-nucleotide polymorphism

4 Clinical Chemistry 65:3 (2019)


Biothreat Applications of Next-Generation Sequencing
Reviews

NGS-based bioforensic applications include out-


break tracing (41– 44 ) and isolate characterization (45 ).
For example, NGS identified causality and traceability in
a hospital outbreak of carbapenem-resistant Klebsiella
pneumoniae and determined how this organism was
transmitted despite rigorous infection-control mecha-
nisms and policies (41 ). NGS linked the index and sub-
sequent cases of K. pneumoniae infection through whole-
genome sequencing characterization and variant analysis.
Epidemiological findings showed the isolate was highly
stable in the hospital environment, thus allowing the bac-
terium to persist on medical equipment despite rigorous
equipment cleaning procedures (41 ). These findings
highlighted the value of NGS in tackling nosocomial
infections through forensic identification of transmission
links and determining gaps in infection-control mea-
sures. Another application of bioforensic NGS is high-
lighted by a study determining the source of a Vibrio
cholerae outbreak in Haiti following the 2010 earth-
quake. Genomic sequencing and other molecular epide-
miological analyses found the outbreak source was likely
Nepalese peacekeepers deployed to Haiti to assist in the
earthquake response (42– 44 ). These data described an
inadvertent, likely single introduction event that resulted
in a rapid outbreak expansion and the first cholera out-
Fig. 3. Timeline of the commercialization of sequencing break in Haiti in approximately 100 years. Lastly, surveil-
platforms alongside instances of notable biological events. lance networks are beginning to use forensic NGS in a
Technological advances in sequencing allowed pathogen charac- broader application of determining transmission of viru-
terization in notable biological events with the exact methods be- lence factors such as antimicrobial resistance. For exam-
ing limited to the technology available at the time. Sanger se- ple, the Multidrug Resistant Organism Repository and
quencing was used with the Amerithrax attacks and NGS in the Surveillance Network used NGS whole-genome se-
West Africa Ebola virus outbreak. Applying sequencing in 1 plat- quencing to identify the first instance of the mcr-13 colis-
form generation or another resulted in characterizing an orga- tin resistance gene in US hospitals (45 ).
nism’s genetic information to better understand the effect and
causality of the biological event. Bioforensic Challenges

All NGS applications struggle with the signal-to-noise or


pathogen-to-background ratio to obtain informative se-
genotypic variants to a specific laboratory-cultivated stock quence information. However, a unique bioforensics
(38 ). Specifically, 4 phenotypic morphotypes in the sample challenge is the requirement for precise assay standard-
were genotypically linked to a parent stock, RMR-1029, ization and validation, as well as chain of custody to meet
resulting in the identification of potential suspects who had legal evidentiary standards and withstand challenge in a
access to this material. Ultimately, the Amerithrax investi- court of law. Validation of techniques must demonstrate
gation led to a nondefinitive conclusion due to circum- the information obtained can answer investigative ques-
stances outside context of forensics-based sequencing tions by assigning the source of a sample while excluding
(39, 40 ). All aspects of the sequence analysis met stringent other sources to the highest degree possible (46, 47 ).
standards of scientific rigor; however, perceived biases in Standard operating procedures are absolutely required as
sample collection, inconsistencies in sampling methods, cul- all components, from sample collection to library prepa-
turing before genetic analysis, and limited sample metadata ration methods, are highly specific to the NGS applica-
contributed to the inability to clearly resolve this case tion. Sequence analysis pipelines including evaluation of
(39, 40 ). The National Biodefense Analysis and Counter- base calling, raw sequence quality, variance analysis, and
measures Center, an agency created for bioforensic applica-
tions, applies lessons learned from the Amerithrax case to the
current application of NGS for bioterrorism and biocrimes
investigations. 3
Gene: mcr-1, mobilized colistin resistance gene.

Clinical Chemistry 65:3 (2019) 5


Reviews

genome assembly also need to be technically validated. ping to the causative agent, Leptospirosis, from a cerebral
This can be a highly onerous process because validation spinal fluid sample. Accomplished within 48 h, this
includes the definition of parameters such as analytical method provided actionable information toward patient
specificity and sensitivity, reproducibility, precision, ac- treatment in a reasonable timeframe, thus allowing incor-
curacy, and robustness (46 ). Unlike many clinical and poration into a standard clinical work flow. However,
biosurveillance samples, forensic samples tend to be the deep sequencing is relatively expensive compared to most
most challenging for NGS because of such factors as lim- diagnostics, effectively pricing its use out of routine clin-
ited sample volume, exposure to and degradation by en- ical applications. In addition, cerebrospinal fluid is a rel-
vironmental elements, and potential mixing with other atively low background clinical specimen compared to
nucleic acid sources (48 ). While challenging, these issues other complex diagnostic matrices. One might expect a
are addressable and are similar to the standard of valida- lower signal-to-noise ratio from whole blood in which
tion required for clinical application and regulatory com- host nucleic acid is at a substantially higher concentra-
pliance of biothreats diagnostics. tions. Issues aside, use of this technology currently runs
the spectrum of applications, with documented use cases
NGS for Biothreat Diagnosis for diagnosis of Zika and Chikungunya virus (53 ), Bru-
cella spp. (54 ), and Ebola virus (28 ) infections.
Clinical applications, perhaps most relevant for this au- Introduction of new technologies into the clinic typ-
dience, are where NGS finds its greatest potential to shift ically follows a path of adoption from Research Use
the diagnostic work flow. NGS infectious diseases diag- Only, to LDT, and finally to Food and Drug Adminis-
nostics are less mature than biosurveillance or bioforen- tration (FDA)-cleared regulatory compliance. Currently,
sics applications; however, NGS is positioned to be a NGS is being adopted as an LDT in several clinical sites
prominent tool in the molecular diagnostic toolbox. As for detection or identification of infectious diseases (55–
mentioned before, NGS requires little to no prior infor- 57 ). Excellent examples of NGS LDT implementation
mation regarding the etiologic agent to identify a patho- include works from the University of Washington
gen or even a mixed population in a culture or sample. (55, 56 ) and Houston Methodist Research Institute
The ability to simultaneously sequence all nucleic acids (57 ). These efforts successfully insert NGS into the clin-
within a sample without analytical specificity is a key ical work flow as a whole-genome characterization tool to
NGS advantage over traditional targeted approaches identify and speciate positive bacterial cultures. Similarly,
such as real-time PCR. However, this lack of specificity is in the biothreat context, efforts to transition research
also a primary obstacle in clinical NGS implementation. methods to the clinic have followed the LDT path
Specifically, in an NGS assay, any nucleic acid in the (50, 58, 59 ). To facilitate the adoption of NGS LDTs,
sample is sequenced (pathogen, commensals, back- the Clinical Laboratory Improvement Amendments
ground biota, and host nucleic acid) and contributes se- (60 ), College of American Pathologists (61 ), and other
quence information to the overall diagnostic output. accrediting bodies (62, 63 ) published roadmaps detail-
Similar to bioforensics and biosurveillance, most clinical ing the validation data sets required to establish an NGS
samples are highly skewed toward host background nu- LDT within the respective clinical laboratory accrediting
cleic acid compared to the pathogen of interest in terms authorities.
of concentration, thus resulting in a poor signal-to-noise
ratio. Numerous groups have found innovative and ele- Regulatory Challenges
gant solutions to this issue including host subtraction and
pathogen enrichment (49 –51 ). These solutions and the Ideally, NGS assays will transition from the LDT status
resulting clinical NGS applications generally focus on 2 to full FDA approval, allowing distribution and use of
primary implementation strategies: diagnostic of last re- these assays in any accredited clinical laboratory without
sort or laboratory-developed tests (LDTs). revalidation. To facilitate this transition, the FDA re-
Wilson et al. efficiently demonstrated the “diagnos- leased draft guidance entitled “Infectious Disease Next
tic of last resort” use case by applying NGS to resolve a Generation Sequencing Based Diagnostic Devices: Mi-
clinical diagnosis when all other diagnostics failed (52 ). crobial Identification and Detection of Antimicrobial
In this study, NGS resolved a cryptic case of leptospirosis Resistance and Virulence Markers” (62 ). This document
that was previously unidentifiable before NGS applica- details a de novo submission for both the modular NGS
tion. Researchers solved the signal-to-noise issue by ap- components FDA considers regulated as well as the ana-
plying high-density sequencing (Illumina HiSeq), pro- lytical and clinical data sets requested for both targeted
viding sufficient coverage to detect the rare pathogen and whole-genome sequencing (Fig. 4). As part of the
reads among substantial human host background (52 ). guidance, FDA defines NGS as a whole system including
Specifically, NGS sequencing produced over 3 million the wet-laboratory component through the bioinformat-
noninformative background reads with only 475 map- ics process to result in a diagnostic output. Perhaps the

6 Clinical Chemistry 65:3 (2019)


Biothreat Applications of Next-Generation Sequencing
Reviews

Fig. 4. Data sets required for an FDA de novo submission for NGS diagnostics.
FDA guidance defines an NGS assay as a whole system from specimen collection through data analysis (blue). All these steps must be
incorporated into the validation studies, generating both the analytical and clinical data sets (gray) required for approval.

most important aspect of this guidance is that it provided gate reagent contamination (66 ). In this context, FDA
a potential relief in the scope of clinical trials, reducing draft guidance addresses the necessity for evaluation of
the data generation burden with a proposed method of in carryover, cross-contamination, and the essential nature
silico, as opposed to wet laboratory, comparison for pos- of nontemplate controls inserted throughout all aspects
itive and negative predictive values. Specifically, assay of sample preparation and library generation. These con-
results from the clinical trial would not need to be com- trols inform the end user of any contamination events in
pared to a gold standard comparator, such as real-time the sequencing reaction, which can later be addressed
PCR or other molecular assay, for every clinical sample bioinformatically with appropriate data analysis.
but instead could be compared to a vetted FDA database
of regulatory-grade sequences (FDA-ARGOS, (64 )). Bioinformatics Considerations
This database is publicly available through NCBI Bio-
project PRJNA231221. NGS for biothreat characterization or clinical identifica-
A substantial regulatory challenge for the clinical tion is only as reliable as the data analysis implemented
adoption of NGS is false-positive reporting due to either after sequencing. There are a number of different se-
nucleic acid contamination of sample processing reagents quencing analysis programs, several of which are tied spe-
or inaccurate index demultiplexing (see section below on cifically to individual sequencing platforms. Others cover
bioinformatics considerations). Nucleic acid reagent the scope of NGS and bioinformatics in broader detail
contamination is common for many molecular biological (67–71 ); however, there are several biothreat-centric
products, e.g., enzymes. Adding steps to the overall sam- considerations for bioinformatics including (a) near-
ple processing, such as extraction and enrichment, adds neighbor differentiation and the implications of false
additional points of potential nucleic acid contamina- positives, (b) biodefense-specific bioinformatics solu-
tion, especially when applying nonspecific amplification tions, and (c) standardization of bioinformatics pipelines
strategies (65 ) and/or ultradeep, unbiased NGS (66 ). to facilitate use of NGS in biothreat-regulated spaces.
Targeted amplification of unique sequences of interest The consequence of misidentifying a sample as a
and physical removal of contaminating nucleic acids via biothreat, when in fact the organism is simply a near
enzymatic, radiation, and chemical treatments can miti- neighbor with a similar genetic composition, can have

Clinical Chemistry 65:3 (2019) 7


Reviews

significant fiscal, logistical, and public perception effects. enomics programs to answer fidelity through consen-
Accurately differentiating a biothreat from near neigh- sus. We have found that both approaches are reliable
bors with a limited number of pathogen reads, typical of but are primarily limited by the scope of current, pub-
unbiased sequencing assays, remains a principal objective licly available sequence databases. As researchers strive
for most biodefense bioinformatics software. Because to push NGS further toward the point of care, the issue
many biothreats are genetically similar to less pathogenic of technical expertise becomes increasing important as most
ancestors, accurately differentiating between the 2 is crit- hospitals and field sites do not have access to or funds to
ically important. For example, a recent report described support trained bioinformatic specialists. Both these soft-
traces of Yersinia pestis and B. anthracis, bacteria that ware programs are potential solutions to this issue.
cause plague and anthrax, respectively, in the New York Standardization of NGS, the wet laboratory, and
subway system (72 ). However, the group and others re- bioinformatics is perhaps the 1 common thread for all
alized after publication that these signatures were not biothreat applications owing to the necessity for regu-
actually specific to these biothreat agents but instead de- lated and validated methods for actionable results. To
rived from phylogenetically similar organisms (73 ). Sim- help standardize NGS, multiple governmental agencies
ilarly, in the process of developing a novel bioinformatics including FDA and National Institutes of Standards and
software, Greninger et al. found Ebola virus reads in an Technology are establishing procedural (64 ) and physi-
influenza case despite the laboratory not having ever pro- cal standards (78 ) for metagenomics that ease the surety
cessed Ebola virus material (74 ). burden placed on computational tools. Using these
Metagenomic sequencing often relies on detection standards, we developed the Diagnostic targETEd
of low numbers of pathogen-specific reads, tens to hun- seQuencing adjudicaTion (DETEQT) software (79 ),
dreds, to make diagnostic calls. As mentioned above, solidifying targeted infectious disease sequencing data
intra- and interassay errors and inaccurate index demul- analysis for LDT applications while minimizing false-
tiplexing can result in false positives due to misassign- discovery rates. In other efforts, FDA and other research-
ment of reads from the same or even previous sequencing ers proposed procedural standards for genome finishing
runs. Intraassay errors stem from hardware and software (80 ) and the generation of high-quality databases, an
issues resulting in misbinning of adjacent clusters into equally important consideration for accurate data analy-
incorrect FASTQ files or misidentification of closely re- sis as assay standardization. Misannotations and poor-
lated indices during demultiplexing (75 ). Interassay con- quality sequences can markedly affect the bioinformatics
tamination can occur on a sequencing run conducted process. Culmination of these standards coupled with
after sequencing a high concentration sample, leading to existing validation procedures will eventually push data-
pathogen-specific read cross-contamination and false- bases and NGS further toward the point of care because
positive results. Given the relatively low number of reads of credible results and ease of use.
used to make a detection call, these issues highlight the
need for assay pipeline validation and confirmatory test- Conclusions
ing for any application in which actionable results would
be required, such as with all NGS biothreat approaches. Over the past 15 years, there have been many technolog-
The biodefense industry created multiple open- ical advancements in biothreat diagnostics and character-
source bioinformatics suites, such as Sequence-based ization. In particular, NGS has greatly expanded the ca-
UltraRapid Pathogen Identification (SURPI) (76 ) and pabilities of detecting biothreats, unexpected pathogens,
Empowering the Development of Genomics Expertise or even the completely novel, previously unknown
(EDGE) (77 ), for highly reliable applications to solve threat. Improvements in cost, throughput, quality,
some the issues of parsing biothreats from near neighbors and time to answer brought NGS from highly special-
and begin to move toward fieldable bioinformatics solu- ized sequencing centers into smaller laboratories, aus-
tions. Though different in execution, these products tere environments, and even the clinic. This broader
strive for the most accurate and reliable answer while NGS availability has enabled many improvements to
minimizing the technical expertise required to run be made across the NGS pipeline, including sample
these software programs. SURPI achieves high result processing, library preparation, and simplified large-
fidelity by liberal sequential removal of noninforma- scale data analysis.
tive sequence reads before matching sequences to As NGS moves into the highly regulated realms of
pathogens. This high fidelity minimizes the potential biothreat diagnostics and bioforensics, substantial efforts
for spurious misassignment of sequence reads, thus will need to be undertaken to meet regulatory and legal
reducing potential for false positives. EDGE ap- requirements, including procedure and bioinformatics
proaches this problem from a different perspective. standardization and validation of sequencing and analysis
After removing host or other problematic signatures, pipelines. Improvements in the technology, specifically
this software uses no fewer than 5 independent metag- with the third generation of sequencers, bring closer to

8 Clinical Chemistry 65:3 (2019)


Biothreat Applications of Next-Generation Sequencing
Reviews

reality the concept of a pathogen-agnostic metagenomic and (d) agreement to be accountable for all aspects of the article thus
diagnostic usable in any location and providing answers ensuring that questions related to the accuracy or integrity of any part of the
within a clinically actionable timeline. This capability article are appropriately investigated and resolved.
will positively affect the public health with better infec- T.D. Minogue, financial support.
tious disease outbreaks response and biosurveillance ef-
Authors’ Disclosures or Potential Conflicts of Interest: Upon man-
forts. In addition, NGS biothreat applications will be a uscript submission, all authors completed the author disclosure form. Dis-
contributing factor for detection and containment of any closures and/or potential conflicts of interest:
biological incident with a high probability of generating
Employment or Leadership: J.W. Koehler, contractor with
attributional sequence information.
USAMRIID.
Consultant or Advisory Role: None declared.
Stock Ownership: None declared.
Author Contributions: All authors confirmed they have contributed to Honoraria: None declared.
the intellectual content of this paper and have met the following 4 require- Research Funding: Defense Threat Reduction Agency project
ments: (a) significant contributions to the conception and design, acquisi- funding.
tion of data, or analysis and interpretation of data; (b) drafting or revising Expert Testimony: None declared.
the article for intellectual content; (c) final approval of the published article; Patents: None declared.

References
1. Rasko DA, Worsham PL, Abshire TG, Stanley ST, Bannan virus origin and transmission during the 2014 out- 24. Permingeat HR, Reggiardo MI, Vallejos RH. Detection
JD, Wilson MR, et al. Bacillus anthracis comparative ge- break. Science 2014;345:1369 –72. and quantification of transgenes in grains by multiplex
nome analysis in support of the Amerithrax investiga- 13. Hoenen T, Safronetz D, Groseth A, Wollenberg KR, Koita and real-time PCR. J Agric Food Chem 2002;50:
tion. Proc Natl Acad Sci USA 2011;108:5027–32. OA, Diarra B, et al. Virology. Mutation rate and geno- 4431– 6.
2. Reich ES. Science after 9/11: how research was changed type variation of Ebola virus from Mali case sequences. 25. Paracchini V, Petrillo M, Reiting R, Angers-Loustau A,
by the September 11 terrorist attacks. https:// Science 2015;348:117–9. Wahler D, Stolz A, et al. Molecular characterization of an
www.scientificamerican.com/article/how-research-was- 14. Sozhamannan S, Holland MY, Hall AT, Negron DA, Ivan- unauthorized genetically modified Bacillus subtilis pro-
changed-by-september-11-terrorist-attacks/ (Accessed cich M, Koehler JW, et al. Evaluation of signature ero- duction strain identified in a vitamin B2 feed additive.
January 2018). sion in Ebola virus due to genomic drift and its impact Food Chem 2017;230:681–9.
3. Rogan DT, Kochar MS, Yang S, Quinn JV. Impact of rapid on the performance of diagnostic assays. Viruses 2015; 26. Willems S, Fraiture MA, Deforce D, De Keersmaecker
molecular respiratory virus testing on real-time decision 7:3130 –54. SC, De Loose M, Ruttink T, et al. Statistical framework for
making in a pediatric emergency department. J Mol Di- 15. Christie A, Davies-Wayne GJ, Cordier-Lassalle T, Black- detection of genetically modified organisms based on
agn 2017;19:460 –7. ley DJ, Laney AS, Williams DE, et al. Possible sexual next generation sequencing. Food Chem 2016;192:
4. Towner JS, Rollin PE, Bausch DG, Sanchez A, Crary SM, transmission of Ebola virus—Liberia, 2015. MMWR 788 –98.
Vincent M, et al. Rapid diagnosis of Ebola hemorrhagic Morb Mortal Wkly Rep 2015;64:479 – 81. 27. Liang C, van Dijk JP, Scholtens IM, Staats M, Prins TW,
fever by reverse transcription-PCR in an outbreak set- 16. Mate SE, Kugelman JR, Nyenswah TG, Ladner JT, Wiley Voorhuijzen MM, et al. Detecting authorized and unau-
ting and assessment of patient viral load as a predictor MR, Cordier-Lassalle T, et al. Molecular evidence of sex- thorized genetically modified organisms containing
of outcome. J Virol 2004;78:4330 – 41. ual transmission of Ebola virus. N Engl J Med 2015; vip3A by real-time PCR and next-generation sequenc-
5. Bausch DG, Rollin PE, Demby AH, Coulibaly M, Kanu J, 373:2448 –54. ing. Anal Bioanal Chem 2014;406:2603–11.
Conteh AS, et al. Diagnosis and clinical virology of Lassa 17. Park DJ, Dudas G, Wohl S, Goba A, Whitmer SL, Ander- 28. Greninger AL, Naccache SN, Federman S, Yu G, Mbala
fever as evaluated by enzyme-linked immunosorbent sen KG, et al. Ebola virus epidemiology, transmission, P, Bres V, et al. Rapid metagenomic identification of
assay, indirect fluorescent-antibody test, and virus iso- and evolution during seven months in Sierra Leone. viral pathogens in clinical samples by real-time nano-
lation. J Clin Microbiol 2000;38:2670 –7. Cell 2015;161:1516 –26. pore sequencing analysis. Genome Med 2015;7:99.
6. Broadhurst MJ, Kelly JD, Miller A, Semper A, Bailey D, 18. Folarin OA, Ehichioya D, Schaffner SF, Winnicki SM, 29. Quick J, Loman NJ, Duraffour S, Simpson JT, Severi E,
Groppelli E, et al. ReEBOV antigen rapid test kit for Wohl S, Eromon P, et al. Ebola virus epidemiology and Cowley L, et al. Real-time, portable genome sequenc-
point-of-care and laboratory-based testing for Ebola vi- evolution in Nigeria. J Infect Dis 2016;214:S102–S9. ing for Ebola surveillance. Nature 2016;530:228 –32.
rus disease: a field validation study. Lancet 2015;386: 19. Lau MS, Dalziel BD, Funk S, McClelland A, Tiffany A, 30. Jeffreys AJ, Brookfield JF, Semeonoff R. Positive iden-
867–74. Riley S, et al. Spatial and temporal dynamics of super- tification of an immigration test-case using human DNA
7. Sanger F, Air GM, Barrell BG, Brown NL, Coulson AR, spreading events in the 2014 –2015 West Africa Ebola fingerprints. Nature 1985;317:818 –9.
Fiddes CA, et al. Nucleotide sequence of bacteriophage epidemic. Proc Natl Acad Sci USA 2017;114:2337– 42. 31. Jeffreys AJ, Wilson V, Thein SL. Individual-specific ‘fin-
phi ×174 DNA. Nature 1977;265:687–95. 20. Holmes EC, Dudas G, Rambaut A, Andersen KG. The gerprints’ of human DNA. Nature 1985;316:76 –9.
8. Sanger F, Nicklen S, Coulson AR. DNA sequencing with evolution of Ebola virus: insights from the 2013–2016 32. Jeffreys AJ, Wilson V, Thein SL. Hypervariable ‘minisat-
chain-terminating inhibitors. Proc Natl Acad Sci USA epidemic. Nature 2016;538:193–200. ellite’ regions in human DNA. Nature 1985;314:
1977;74:5463–7. 21. Noyce RS, Lederman S, Evans DH. Construction of an 67–73.
9. Heather JM, Chain B. The sequence of sequencers: the infectious horsepox virus vaccine from chemically 33. Roewer L. DNA fingerprinting in forensics: past, pres-
history of sequencing DNA. Genomics 2016;107:1– 8. synthesized DNA fragments. PLoS One 2018;13: ent, future. Investig Genet 2013;4:22.
10. Grard G, Fair JN, Lee D, Slikas E, Steffen I, Muyembe JJ, e0188453. 34. Yang Y, Xie B, Yan J. Application of next-generation se-
et al. A novel rhabdovirus associated with acute hemor- 22. Kerr PJ, Perkins HD, Inglis B, Stagg R, McLaughlin E, quencing technology in forensic science. Genomics
rhagic fever in central Africa. PLoS Pathog 2012;8: Collins SV, Van Leeuwen BH. Expression of rabbit IL-4 Proteomics Bioinformatics 2014;12:190 –7.
e1002924. by recombinant myxoma viruses enhances virulence 35. Torok TJ, Tauxe RV, Wise RP, Livengood JR, Sokolow R,
11. Briese T, Paweska JT, McMullan LK, Hutchison SK, and overcomes genetic resistance to myxomatosis. Vi- Mauvais S, et al. A large community outbreak of salmo-
Street C, Palacios G, et al. Genetic detection and charac- rology 2004;324:117–28. nellosis caused by intentional contamination of restau-
terization of Lujo virus, a new hemorrhagic fever- 23. Hernandez M, Pla M, Esteve T, Prat S, Puigdomenech P, rant salad bars. JAMA 1997;278:389 –95.
associated arenavirus from southern Africa. PLoS Pat- Ferrando A. A specific real-time quantitative PCR detec- 36. Metzker ML, Mindell DP, Liu XM, Ptak RG, Gibbs RA,
hog 2009;5:e1000455. tion system for event MON810 in maize yieldgard Hillis DM. Molecular evidence of HIV-1 transmission in
12. Gire SK, Goba A, Andersen KG, Sealfon RS, Park DJ, based on the 3`-transgene integration sequence. Trans- a criminal case. Proc Natl Acad Sci USA 2002;99:
Kanneh L, et al. Genomic surveillance elucidates Ebola genic Res 2003;12:179 – 89. 14292–7.

Clinical Chemistry 65:3 (2019) 9


Reviews

37. Hoffmaster AR, Fitzgerald CC, Ribot E, Mayer LW, Pop- Grant P, et al. Specific capture and whole-genome se- 65. Salter SJ, Cox MJ, Turek EM, Calus ST, Cookson WO,
ovic T. Molecular subtyping of Bacillus anthracis and quencing of viruses from clinical samples. PLoS One Moffatt MF, et al. Reagent and laboratory contamina-
the 2001 bioterrorism-associated anthrax outbreak, 2011;6:e27805. tion can critically impact sequence-based microbiome
United States. Emerg Infect Dis 2002;8:1111– 6. 52. Wilson MR, Naccache SN, Samayoa E, Biagtan M, analyses. BMC Biol 2014;12:87.
38. Read TD, Salzberg SL, Pop M, Shumway M, Umayam L, Bashir H, Yu G, et al. Actionable diagnosis of neurolep- 66. Laurence M, Hatzis C, Brash DE. Common contami-
Jiang L, et al. Comparative genome sequencing for dis- tospirosis by next-generation sequencing. N Engl nants in next-generation sequencing that hinder dis-
covery of novel polymorphisms in Bacillus anthracis. J Med 2014;370:2408 –17. covery of low-abundance microbes. PLoS One 2014;9:
Science 2002;296:2028 –33. 53. Sardi SI, Somasekar S, Naccache SN, Bandeira AC, Tauro e97876.
39. Gilchrist CA, Turner SD, Riley MF, Petri WA, Jr., Hewlett LB, Campos GS, Chiu CY. Coinfections of Zika and Chi- 67. Valdivia-Granda WA. Biodefense oriented genomic-
EL. Whole-genome sequencing in outbreak analysis. kungunya viruses in Bahia, Brazil, identified by metag- based pathogen classification systems: challenges and
Clin Microbiol Rev 2015;28:541– 63. enomic next-generation sequencing. J Clin Microbiol opportunities. J Bioterror Biodef 2012;3:1000113.
40. Committee on Review of the Scientific Approaches 2016;54:2348 –53. 68. Karlsson OE, Hansen T, Knutsson R, Lofstrom C, Gran-
Used During the FBI’s Investigation of the 2001 Bacillus 54. Mongkolrattanothai K, Naccache SN, Bender JM, Sa- berg F, Berg M. Metagenomic detection methods in
anthracis mailings. Review of the scientific approaches mayoa E, Pham E, Yu G, et al. Neurobrucellosis: unex- biopreparedness outbreak scenarios. Biosecur Bioter-
used during the FBI’s investigation of the 2001 anthrax pected answer from metagenomic next-generation se- ror 11 Suppl 1:S146 –57, 2013.
letters. Washington (DC): National Academies Press; quencing. J Pediatric Infect Dis Soc 2017;6:393– 8. 69. Minogue TD, Koehler JW, Norwood DA. Targeted next-
2011. 55. Roach DJ, Burton JN, Lee C, Stackhouse B, Butler-Wu generation sequencing for diagnostics and forensics.
41. Snitkin ES, Zelazny AM, Thomas PJ, Stock F, Group SM, Cookson BT, et al. Correction: a year of infection in Clin Chem 2017;63:450 –2.
NCSP, Henderson DK, et al. Tracking a hospital out- the intensive care unit: prospective whole genome se- 70. Fricke WF, Rasko DA, Ravel J. The role of genomics in
break of carbapenem-resistant Klebsiella pneumoniae quencing of bacterial clinical isolates reveals cryptic the identification, prediction, and prevention of biolog-
with whole-genome sequencing. Sci Transl Med 2012; transmissions and novel microbiota. PLoS Genet 2017; ical threats. PLoS Biol 2009;7:e1000217.
4:148ra16. 13:e1006724. 71. Greene JM, Collins F, Lefkowitz EJ, Roos D, Scheuer-
42. Eppinger M, Pearson T, Koenig SS, Pearson O, Hicks N, 56. Salipante SJ, Sengupta DJ, Rosenthal C, Costa G, Span- mann RH, Sobral B, et al. National Institute of Allergy
Agrawal S, et al. Genomic epidemiology of the Haitian gler J, Sims EH, et al. Rapid 16S rrna next-generation
and Infectious Diseases bioinformatics resource
cholera outbreak: a single introduction followed by sequencing of polymicrobial clinical samples for diag-
centers: new assets for pathogen informatics. Infect Im-
rapid, extensive, and continued spread characterized nosis of complex bacterial infections. PLoS One 2013;
mun 2007;75:3212–9.
the onset of the epidemic. MBio 2014;5:e01721. 8:e65226.
72. Afshinnekoo E, Meydan C, Chowdhury S, Jaroudi D,
43. Hendriksen RS, Price LB, Schupp JM, Gillece JD, Kaas 57. Long SW, Williams D, Valson C, Cantu CC, Cernoch P,
Boyer C, Bernstein N, et al. Geospatial resolution of hu-
RS, Engelthaler DM, et al. Population genetics of Vibrio Musser JM, Olsen RJ. A genomic day in the life of a
man and bacterial diversity with city-scale metagenom-
cholerae from Nepal in 2010: evidence on the origin of clinical microbiology laboratory. J Clin Microbiol 2013;
ics. Cell Syst 2015;1:97– e3.
the Haitian outbreak. MBio 2011;2:e00157–11. 51:1272–7.
73. Ackelsberg J, Rakeman J, Hughes S, Petersen J, Mead
44. Chin CS, Sorenson J, Harris JB, Robins WP, Charles RC, 58. Stefan CP, Koehler JW, Minogue TD. Targeted next-
P, Schriefer M, et al. Lack of evidence for plague or an-
Jean-Charles RR, et al. The origin of the Haitian cholera generation sequencing for the detection of ciprofloxa-
thrax on the New York city subway. Cell Syst 2015;1:
outbreak strain. N Engl J Med 2011;364:33– 42. cin resistance markers using molecular inversion
4 –5.
45. McGann P, Snesrud E, Maybank R, Corey B, Ong AC, probes. Sci Rep 2016;6:25904.
74. Greninger AL, Chen EC, Sittler T, Scheinerman A, Rou-
Clifford R, et al. Escherichia coli harboring mcr-1 and 59. Stefan CP, Hall AT, Minogue TD. Detection of 16S rRNA
binian N, Yu G, et al. A metagenomic analysis of pan-
blaCTX-M on a novel IncF plasmid: first report of mcr-1 and KPC genes from complex matrix utilizing a molec-
demic influenza a (2009 H1N1) infection in patients
in the United States. Antimicrob Agents Chemother ular inversion probe assay for next-generation se-
2016;60:4420 –1. quencing. Sci Rep 2018;8:2028. from North America. PLoS One 2010;5:e13381.
46. Budowle B, Schutzer SE, Morse SA, Martinez KF, 60. Gargis AS, Kalman L, Berry MW, Bick DP, Dimmock DP, 75. Kircher M, Sawyer S, Meyer M. Double indexing over-
Chakraborty R, Marrone BL, et al. Criteria for validation Hambuch T, et al. Assuring the quality of next- comes inaccuracies in multiplex sequencing on the Il-
of methods in microbial forensics. Appl Environ Micro- generation sequencing in clinical laboratory practice. lumina platform. Nucleic Acids Res 2012;40:e3.
biol 2008;74:5599 – 607. Nat Biotechnol 2012;30:1033– 6. 76. Naccache SN, Federman S, Veeraraghavan N, Zaharia
47. Budowle B, Connell ND, Bielecka-Oder A, Colwell RR, 61. Aziz N, Zhao Q, Bry L, Driscoll DK, Funke B, Gibson JS, et M, Lee D, Samayoa E, et al. A cloud-compatible bioin-
Corbett CR, Fletcher J, et al. Validation of high through- al. College of American pathologists’ laboratory stan- formatics pipeline for ultrarapid pathogen identifica-
put sequencing and microbial forensics applications. dards for next-generation sequencing clinical tests. tion from next-generation sequencing of clinical sam-
Investig Genet 2014;5:9. Arch Pathol Lab Med 2015;139:481–93. ples. Genome Res 2014;24:1180 –92.
48. Kuiper I. Microbial forensics: next-generation sequenc- 62. United States Food and Drug Administration. Draft 77. Li PE, Lo CC, Anderson JJ, Davenport KW, Bishop-Lilly
ing as catalyst: the use of new sequencing technologies guidance: infectious disease next generation sequenc- KA, Xu Y, et al. Enabling the democratization of the
to analyze whole microbial communities could become ing based diagnostic devices: microbial identification genomics revolution with a fully integrated web-based
a powerful tool for forensic and criminal investigations. and detection of antimicrobial resistance and virulence bioinformatics platform. Nucleic Acids Res 2017;45:
EMBO Rep 2016;17:1085–7. markers. https://www.fda.gov/downloads/Medical 67– 80.
49. Gardner SN, Frey KG, Redden CL, Thissen JB, Allen JE, Devices/DeviceRegulationandGuidance/Guidance 78. Mason CE, Afshinnekoo E, Tighe S, Wu S, Levy S. Inter-
Allred AF, et al. Targeted amplification for enhanced Documents/UCM500441.pdf (Accessed Jan. 2013). national standards for genomes, transcriptomes, and
detection of biothreat agents by next-generation se- 63. Gargis AS, Kalman L, Lubin IM. Assuring the quality of metagenomes. J Biomol Tech 2017;28:8 –18.
quencing. BMC Res Notes 2015;8:682. next-generation sequencing in clinical microbiology 79. Conrad TA. Diagnostic targeted sequencing adjudication
50. Koehler JW, Hall AT, Rolfe PA, Honko AN, Palacios GF, and public health laboratories. J Clin Microbiol 2016; (deteqt). https://www.mathworks.com/matlabcentral/
Fair JN, et al. Development and evaluation of a panel of 54:2857– 65. fileexchange/66892-deteqt.
filovirus sequence capture probes for pathogen detec- 64. Sichtig H. Infectious disease next generation sequenc- 80. Chain PS, Grafham DV, Fulton RS, Fitzgerald MG,
tion by next-generation sequencing. PLoS One 2014;9: ing based diagnostic devices: microbial identification Hostetler J, Muzny D, et al. Genomics. Genome project
e107007. and detection of antimicrobial resistance and virulence standards in a new era of sequencing. Science 2009;
51. Depledge DP, Palser AL, Watson SJ, Lai IY, Gray ER, markers. FDA; 2016. 326:236 –7.

10 Clinical Chemistry 65:3 (2019)

You might also like