ArticlePDF Available

Ebola Virus Diagnostics: The US Centers for Disease Control and Prevention Laboratory in Sierra Leone, August 2014 to March 2015

Authors:

Abstract and Figures

In August 2014, the Viral Special Pathogens Branch of the US Centers for Disease Control and Prevention established a field laboratory in Sierra Leone in response to the ongoing Ebola virus outbreak. Through March 2015, this laboratory tested >12 000 specimens from throughout Sierra Leone. We describe the organization and procedures of the laboratory located in Bo, Sierra Leone. Published by Oxford University Press on behalf of the Infectious Diseases Society of America 2015. This work is written by (a) US Government employee(s) and is in the public domain in the US.
Content may be subject to copyright.
SUPPLEMENT ARTICLE
Ebola Virus Diagnostics: The US Centers for
Disease Control and Prevention Laboratory in
Sierra Leone, August 2014 to March 2015
Mike Flint,1Christin H. Goodman,14 Scott Bearden,15 Dianna M. Blau,2Brian R. Amman,1Alison J. Basile,14
Jessica A. Belser,3Éric Bergeron,1Michael D. Bowen,4Aaron C. Brault,14 Shelley Campbell,1Ayan K. Chakrabarti,1
Kimberly A. Dodd,16 Bobbie R. Erickson,1Molly M. Freeman,5Aridth Gibbons,1Lisa W. Guerrero,1John D. Klena,17
R. Ryan Lash,6Michael K. Lo,1Laura K. McMullan,1Gbetuwa Momoh,18 James L. Massally,18 Augustine Goba,18
Christopher D. Paddock,7Rachael A. Priestley,7Meredith Pyle,8Mark Rayfield,9Brandy J. Russell,14 Johanna S. Salzer,10
Angela J. Sanchez,13 Amy J. Schuh,1Tara K. Sealy,1Martin Steinau,11 Robyn A. Stoddard,12 Céline Taboy,1
Maryann Turnsek,5David Wang,3Galina E. Zemtsova,7Marko Zivcec,1Christina F. Spiropoulou,1Ute Ströher,1
Jonathan S. Towner,1Stuart T. Nichol,1and Brian H. Bird1
1
Viral Special Pathogens,
2
Infectious Diseases Pathology,
3
Influenza Division, Immunology and Pathogenesis,
4
Gastroenteritis and Respiratory Virus
Laboratory,
5
Enteric Diseases Laboratory,
6
TravelersHealth,
7
Rickettsial Zoonoses,
8
Laboratory Research,
9
Global Disease Detection,
10
Poxvirus and
Rabies,
11
Chronic Viral Diseases,
12
Bacterial Special Pathogens Branches, and
13
Office of Technology and Innovation, Centers for Disease Control and
Prevention, Atlanta, Georgia;
14
Arboviral Diseases, and
15
Bacterial Diseases branches, Centers for Disease Control and Prevention, Fort Collins, Colorado;
16
University of California, Davis, School of Veterinary Medicine;
17
Division of Global Health Protection, Centers for Disease Control and Prevention, Beijing,
China; and
18
Ministry of Health and Sanitation, Kenema Government Hospital, Sierra Leone
In August 2014, the Viral Special Pathogens Branch of the US Centers for Disease Control and Prevention
established a eld laboratory in Sierra Leone in response to the ongoing Ebola virus outbreak. Through
March 2015, this laboratory tested >12 000 specimens from throughout Sierra Leone. We describe the
organization and procedures of the laboratory located in Bo, Sierra Leone.
Keywords.Ebola; diagnostics; eld laboratory.
Since the discovery of Ebola virus (EBOV) in 1976,
there have been >20 outbreaks, but the West African
epidemic of 20132015 is the largest recorded, with
>10 000 fatalities and 25 000 persons infected as of 29
March 2015, surpassing all the other outbreaks com-
bined. EBOV is classied in the family Filoviridae,
genus Ebolavirus,withtheZaire ebolavirus species
being responsible for the 2014 West African outbreak
[1]. The EBOV genome is negative-strand RNA approx-
imately 19 kb long that encodes 7 genes: nucleoprotein
(NP), viral protein (VP) 35, VP40, glycoprotein, VP30,
VP24 and the viral polymerase L [2].Several tests can be
used to diagnose EBOV infection, including reverse-
transcription polymerase chain reaction (RT-PCR) to
detect the virus genome [36], enzyme-linked immuno-
sorbent assay to measure virus-specic immunoglobu-
lin (Ig) M or IgG [7,8], or assays for viral antigen, either
in blood (with enzyme-linked immunosorbent assay)
[7] or in skin or liver biopsy specimens (with immuno-
histochemistry) [9,10]. RT-PCR has been the most
common assay used in the current outbreak, detecting
EBOV RNA in whole blood samples obtained from liv-
ing patients or oral swab samples collected from dead
bodies.
Laboratories have been established in many parts of
the outbreak region, often associated with an EBOV
treatment unit (ETU), to provide testing for diagnostic
purposes, aiding admittance, treatment, and discharge
decisions. For living patients, RT-PCR may give a neg-
ative result if performed <72 hours after onset of clinical
symptoms, apparently owing to low viremia during the
early stages of infection [11,12]. Thus, a negative test
result may be valid only if the blood sample is obtained
Correspondence: Stuart T. Nichol, Viral Special Pathogens Branch, Division of
High Consequence Pathogens and Pathology, National Center for Emerging and Zoo-
notic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton
Rd, Mail Stop G-14, Atlanta, GA 30333 (stn1@cdc.gov).
The Journal of Infectious Diseases
®
Published by Oxford University Press on behalf of the Infectious Diseases Society of
America 2015. This work is written by (a) US Government employee(s) and is in the
public domain in the US.
DOI: 10.1093/infdis/jiv361
Ebola Field Laboratory JID S1
Journal of Infectious Diseases Advance Access published July 30, 2015
at Stephen B. Thacker CDC Library on August 4, 2015http://jid.oxfordjournals.org/Downloaded from
72 hours after onset. Because reverse-transcriptase inhibitors
in the sample can cause false-negative results, the laboratory will
usually include a control for this, either through amplication
of a human transcript such as β2-microglobulin (B2M; which
also acts as a check for sample integrity), or by addition of an
exogenous RNA, which is then also detected with RT-PCR.
The Viral Special Pathogens Branch (VSPB) of the US Cen-
ters for Disease Control and Prevention (CDC) has many de-
cades of experience in establishing and operating diagnostic
laboratories in low-resource settings, in support of public health
efforts to control EBOV and Marburg virus disease outbreaks.
In August 2014, in response to an ongoing EBOV outbreak in
West Africa, the Sierra Leone Ministry of Health and Sanitation
invited the CDC to establish an EBOV diagnostic laboratory in
Sierra Leone. The laboratory was initially situated at Kenema
Government Hospital, but owing to westward movement of
cases, it was moved in late September to the Bandajuma region
of Bo within the Médicins Sans Frontières (MSF) case manage-
ment center (Figure 1). When it opened, the CDC laboratory
was one of only 3 EBOV diagnostic laboratories operating in Si-
erra Leone, and during the next 7 months it received samples
from 12 of Sierra Leones 14 districts. Here we describe the or-
ganization and work ow of the Bo laboratory. We hope this in-
formation will facilitate the establishment and aid the rapid
deployment of EBOV diagnostic eld laboratories for this and
future outbreaks.
MATERIALS AND METHODS
Biosafety
It was not possible to establish a biosafety level 4 (BSL-4) con-
tainment laboratory in Sierra Leone. To allow CDC scientists to
work safely, a hot laboratorywas set up. This was the only area
where patient samples were handled. All persons entering the
hot laboratory were trained by VSPB scientists experienced in
working with EBOV under BSL-4 conditions. The personal pro-
tective equipment (PPE) worn in the hot laboratory included
scrubs and Croc clog shoes (Uniform Advantage), level 3 surgi-
cal gowns (Proxima Sirus; Medline Industries), gloves (High
Five Products), extended cuff gloves (Purple Nitrile-Xtra; Kim-
berly-Clark), and shoe covers (Proshield 3; DuPont). Powered
air purifying respirators (PAPRs) and accessories were manu-
factured by 3M (Air-Mate). Potentially infected materials
were decontaminated using 5% (vol/vol) Micro-Chem Plus dis-
infectant: a mixture of dimethyl benzyl ammonium chloride,
dimethyl ethyl benzyl ammonium chloride, and polyethylene
mono-ether glycols (National Chemical Laboratories). Extensive
safety testing conrmed that this concentration of disinfectant
was virucidal for EBOV (P. Jahrling, personal communication).
Specimen transport containers were Air Sea BioJar (code 500;
Biopack 2; 1.5-L UN combination packaging 4G/class 6.2)
from Air Sea Containers, and absorbent paper pads were from
Saf-T-Pak.
RNA Extraction and RT-PCR Assays
RNA was extracted from patient specimens using the MagMAX
magnetic bead system (Life Technologies), according to the
manufacturers instructions. In this process, 100 µL of each
specimen was mixed with 400 µL of lysis buffer supplemented
with 2 µL of carrier RNA solution. RNA was extracted, using
either a MagMAX Express-96 Deep-Well magnetic particle ex-
tractor or a BeadRetriever automated magnetic bead separation
system, and then eluted in 90 µL of elution buffer. Full details of
the RT-PCR assays will be described elsewhere, but are available
on request. Briey, 3 tests were performed concurrently: 2 spe-
cic for EBOV NP and VP40 genes and 1 for the human B2M
gene as a control for sample integrity. RT-PCR assays were per-
formed on a CFX96 Real-Time System (BioRad). Samples for
use as positive controls were generated in the BSL-4 laboratory
at the CDC in Atlanta, Georgia; Z. ebolavirus (Mayinga variant
from the 1976 outbreak) from the VSPB collection was grown in
Vero E6 cells, and viral RNA was extracted from the culture
medium.
Results from the quantitative RT-PCR (qRT-PCR) assays
were evaluated comprehensively with attention paid to each
of the 3 targets qRT-PCR test results, the date of onset of clinical
Figure 1. Map of Sierra Leone, with the different districts shown. Dis-
trict names are in black text, city names in white. The Centers for Disease
Control and Prevention laboratory was initially established in Kenema but
was relocated to Bo in September 2014.
S2 JID Flint et al
at Stephen B. Thacker CDC Library on August 4, 2015http://jid.oxfordjournals.org/Downloaded from
illness, the date of collection of the specimen, and the case his-
tory of the patient (if known). Specimens were considered neg-
ative if 3 criteria were met: (1) both EBOV NP and VP40 were
undetected, (2) the internal extraction control B2M was detect-
ed in an acceptable range of cycle threshold values (blood spec-
imens, approximately 1830; oral swab samples, approximately
2839), and (3) the specimen was collected >72 hours after
onset of clinical illness. Specimens were considered positive re-
gardless of the timing of collection relative to onset of clinical
illness if (1) either of the NP or VP40 assays were detected
(cycle threshold, <39) and (2) the B2M internal control was de-
tected in the acceptable ranges. In instances where results were
discordant between the NP and VP40 assays (typically in the
rst 13 days after onset of illness when viremia levels were
lower) a follow-up specimen was requested for nal denitive
conrmation.
Results were considered pending if (1) the NP and VP40 as-
says were not detected and the B2M internal control also failed
detection or (2) the specimen was collected <72 hours after
onset of clinical illness. In these pending cases, a subsequent
(>72 hours) follow-up specimen was requested for denitive di-
agnosis of EBOV status. A small minority of specimens were re-
jected as nondiagnostic if the B2M internal control failed to be
detected after 2 separate RNA extractions and qRT-PCR test
runs. The overwhelming majority of the rejected specimens
were surveillance oral swab samples obtained from dead bodies
whose poor quality was probably due either to improper collec-
tion technique or to excessive transit time to the laboratory.
RESULTS
Establishment of the CDC Field Laboratory in Sierra Leone for
EBOV Diagnostics
MSF provided a building on the grounds of their Bo case man-
agement center for CDC use; this space was converted into a
eld laboratory. Running water was provided by the MSF
water and sanitation engineers, and electric power was supplied
by medium capacity generators on the MSF compound and
with small portable high quality backup generators (Hon-
daEU2000i) for sensitive electronic equipment. Three rooms
within the house were designated as work areas: a clean room
to set up PCR and RNA extraction reagents, a room for extract-
ing RNA and adding it to PCR plates, and a room for perform-
ing the PCR assays (Figure 2A). The third room also served as
an ofce area for laboratory workers during their deployment.
Although many eld laboratories opt to use class III biolog-
ical safety cabinets (glove boxes), we established a separate hot
laboratory. Glove boxes require electrical power, can be uncom-
fortable to work in for extended periods, and the glove ports can
block the scientistseld of view while manipulating samples.
The gloves may be corroded and weakened by repeated expo-
sure to the decontaminating solution. However, they can be
used with a more minimal PPE and in the presence of air con-
ditioning. In contrast, a hot laboratory, with a larger working
area and less restricted movement, allows multiple scientists
to work on the same specimen, in a production line with differ-
ent scientists performing individual steps of the processing
sequence.
For the hot laboratory, MSF constructed a wood-framed scaf-
fold with corrugated iron roong and plastic sheeting walls
about 20 yards from the main laboratory building (Figure 2B).
An anteroom was used as a changing area where scrubs, PPE,
and PAPRs were donned and doffed. To allow transfer of ma-
terials out of the hot laboratory, a storage trunk was lled with
disinfectant (approximately 40 L) and situated with one end ac-
cessible from the hot area and the other end in the changing
room. This was used as a dunk tank,in which items from
the hot area could be submerged and the exterior surfaces dis-
infected before retrieval in the changing room. The disinfectant
solution in the dunk tank was changed once every 34 weeks.
Only a single area (room 1 inside the house) was air condi-
tioned, and temperatures in the other rooms, especially the hot
lab, frequently exceeded 32°C. Consequently, when workers
were inside the hot laboratory, those outside would check on
their status every 510 minutes. In addition, a wireless doorbell
system was arranged to allow signaling from inside the hot lab-
oratory to the outside, to indicate that assistance was required.
The hot laboratory was not a closed structure, having an open-
ing between the walls and the roong all around its perimeter
(Figure 2B), so it was possible to communicate with those inside
from the anteroom area. For routine communications with sci-
entists working in the hot laboratory no additional PPE was re-
quired for those in the anteroom.
The CDC laboratory teams routinely consisted of 4 members:
3 scientists to perform sample testing and a team leader respon-
sible for data entry, quality control, and reporting of results. On
days with high sample loads, 2 scientists processed samples in
the hot laboratory, while the other scientist performed RNA ex-
traction and PCR in the main laboratory building. On days with
fewer samples, a single person was required for the hot labora-
tory, and the other 2 implemented RNA extraction and PCR.
On most days, a morning and an afternoon extraction and
RT-PCR assay were performed. Generally, the laboratory
teams were deployed for 28 days, with 13daysofoverlap
with the previous team. Each team worked 1014 hours a
day, 7 days a week, with no days off.
Daily Work Flow and Specimen Processing
Samples were received from referring facilities, most frequently
delivered by a motorcycle courier, but sometimes delivered by
ambulance or by a United Nationscoordinated helicopter.
Most deliveries, but not all, were accompanied by the appropri-
ate case investigation forms. On receipt of the sample, the trans-
port boxes, the outside of the triple-package Air Sea shipping
Ebola Field Laboratory JID S3
at Stephen B. Thacker CDC Library on August 4, 2015http://jid.oxfordjournals.org/Downloaded from
containers, and each sheet of paperwork were decontaminated
by misting with disinfectant. The sheets of paper were allowed
to air dry before examination. The containers were stored, un-
opened, at 4°C before processing and were never opened outside
the hot laboratory. For each sample listed on the case investiga-
tion form, the time of delivery, the patients name, and the re-
ferring facilitysidentication number was recorded, and a
unique laboratory identication number was assigned. A 2-
mL Wheaton cryogenic tube and a screw-cap Sarstedt tube
were labeled with the same unique laboratory identier, the for-
mer for an aliquot of the sample material (for freezing, in case a
repeat of the assay was required), the latter tube with 400 µL of
lysis buffer supplemented with carrier RNA for sample inacti-
vation before RNA extraction.
Owing to differences in work requirements, the PPE used in
the hot laboratory differed signicantly from that used by MSF
clinicians for patient care in treatment centers (Figure 3). The
hot laboratory workers used lightweight level 3 laboratory
gowns, which are much cooler than a full-Tyvek suit, an impor-
tant consideration given the ambient environmental conditions.
For face and respiratory protection instead of N-95 respirators
and goggles, the hot laboratory workers wore a full-face shield
and hooded PAPR, which, in addition to respiratory protection,
provided some air ow and relief from the heat. The lter unit
of the PAPR was worn underneath the gowns and was fully pro-
tected from contamination, thus allowing for easier surface de-
contamination without harming the internal high-efciency
particulate air lter and electronics.
In the changing room of the hot laboratory structure, scien-
tists removed all jewelry and clothing, including underwear, and
put on scrubs, socks, and lightweight plastic footwear (Crocs or
equivalent) and shoe covers. The scrub pants were tucked into
the socks and taped in place to prevent them from falling and
revealing the skin. A PAPR with full hood was donned next,
Figure 2. A, Floor plan of the Centers for Disease Control and Prevention (CDC) laboratory at Bo. Room 1 is the clean room, used for preparation of
polymerase chain reaction (PCR) master mix and reagents for RNA extraction, room 2 is used for RNA extraction and the addition of RNA to PCR plates, and
room 3 is an office area with thermocyclers, used for performing the PCR reactions and for data analysis. Work is undertaken in a unidirectional flow, from
room 1 to room 2 to room 3.
S4 JID Flint et al
at Stephen B. Thacker CDC Library on August 4, 2015http://jid.oxfordjournals.org/Downloaded from
with a level 3 closed-front gown over the scrubs with the upper
portion of the gown tting between the inner and outer folds of
the PAPR hood. An inner pair of gloves was taped to the sleeves
of the gown; a middle pair, with extended cuffs, went over the
rst pair and was taped also. A third pair of gloves went over the
second pair, the third pair being light colored to make any
splashesontheglovesmorevisible.Oncedonned,thePPE
was checked by another scientist, with special attention to the
rear of the gown to ensure that the PAPR and the scientists
back were not exposed.
The objective of the hot laboratory work was to ensure that
the received samples were correctly documented, to transfer an
aliquot of the specimen to a cryogenic tube for freezing, and to
place 100 µL of the specimen into lysis buffer for RNA extrac-
tion. Scientists therefore entered the hot laboratory with the
shipping containers, a list of the samples that were documented
as being inside the containers, cryovials for specimen storage,
and tubes containing lysis buffer (each tube labeled with a
unique VSPB identifying number). The rst task inside the
hot laboratory was to open the shipping containers and to
sort the specimens for processing, identify any that might be
missing or unlisted, and record any information present on
the specimen containers that differed from that on the case in-
vestigation form. A scientist outside the hot laboratory took
notes of discrepancies, passed in additional tubes, and corrected
the log books if necessary.
Once sample identication and documentation was com-
plete, the specimen tubes were opened one at a time at arms
length over a bucket of disinfectant. When possible, a dispos-
able transfer pipette was used to transfer up to 1.5 mL of the
sample to the 2-mL cryogenic tube. Specimens that were inac-
cessible by transfer pipette were retrieved using a regular micro-
pipette with an aerosol barrier tip. Then, from the cryogenic
tube, 100 µL of specimen was transferred to a screw-cap tube
containing lysis buffer and carrier RNA. The latter tube was
shaken to ensure complete mixing, and each tube was trans-
ferred to a plastic, lidded box with holes in the bottom (so
that the box would ood with disinfectant when submerged
in the dunk tank, without the tubes oating out). After use,
each pipette tip or transfer pipette was used to aspirate disinfec-
tant to decontaminate its interior, and then it was discarded
into a disinfectant waste bucket, along with any excess specimen
and the now empty specimen tubes. After processing of each
sample, the scientists gloves were sprayed with disinfectant. If
Figure 2 continued.B,Hot laboratoryat the CDC Bo laboratory. Constructed by Médicins Sans Frontières, this is a temporary wooden structure with
corrugated metal roofing and plastic sheeting walls. An anteroom serves as a changing area, separated from the exterior by a plastic sheet that can be
raised or lowered for privacy. Abbreviation: PPE, personal protective equipment.
Ebola Field Laboratory JID S5
at Stephen B. Thacker CDC Library on August 4, 2015http://jid.oxfordjournals.org/Downloaded from
pipettors were used, they were wiped down with a paper towel
soaked in disinfectant after each sample.
Once all samples were processed, the plastic boxes were im-
mersed in the dunk tank, and weighed down with a rock, for at
least a 3-minute contact time. The reusable shipping containers
were dunked outin the same fashion. To leave the hot labo-
ratory, the scientist opened the door back to the changing room
and, while standing in the doorway, was sprayed from head to
toe with disinfectant by another scientist wearing a face shield,
gown, and gloves. Once sprayed, the hot laboratory scientist re-
moved and discarded his or her shoe covers and outer gloves.
The shoes, middle gloves, and tape attaching the gloves to the
gown were also then sprayed with disinfectant, and the middle
gloves removed. The inner gloves were then sprayed with dis-
infectant, and the scientists stepped out of the hot area. The
tape, the gown, and the inner gloves were removed and safely
discarded. The PAPR hood was removed, with only the interior
surface touched. Tubes were retrieved from the dunk tank,
specimen aliquots were frozen at 20°C, and RNA was extract-
ed from the samples in lysis buffer.
Discarded specimen tubes, transfer pipettes, and tips were
soaked in disinfectant overnight, before disposal the next morn-
ing. A colander set over a bucket was used to lter them from
the liquid, and they were then disposed along with other solid
waste, double bagged in biohazard autoclave bags. The exterior
of the double-bagged trash was sprayed with disinfectant, trans-
ferred out of the hot laboratory, and placed in a third biohazard
bag, which was in turn decontaminated. For liquid waste, the
lid, handle, surfaces and sides of the waste bucket were sprayed
with disinfectant, and the bottom of the bucket was dipped into
the dunk tank. The bucket could then be passed out of the hot
laboratory. Solid waste was incinerated by MSF workers, and
liquid waste was disposed in the compound septic tank.
Challenges and Solutions
The packaging of samples delivered to the laboratory was fre-
quently challenging. Samples were generally either whole blood
obtained from living patients, into purple-top tubes containing
anticoagulant ethylenediaminetetraacetic acid or red-top tubes
with no anticoagulant, or oral swab samples obtained from
dead bodies (in a transport medium). The supply chain to the
districts was inconsistent for several months; initially, samples
were received in a variety of containers, including coffee pots
(possibly intended to insulate the samples), bloodied urine con-
tainers wrapped in gloves, lled syringes with needles attached,
and sample tubes in plastic shopping bags. Occasionally samples
were received many days after collection, with the clotted blood
stuck to the stopper lids. The referring facilities, when it was
possible to contact them, were responsive to feedback and were
generally able to work toward xing these biosafety issues. To im-
prove sample transport safety, materials for a triple-layer packing
system were purchased by CDC and provided to couriers on each
sample delivery for return to the referring facilities. The packing
system consisted of a watertight, leak-proof, shatter-resistant
shipping container lled with soft-paper tissues for padding
and reclosable bags containing absorbent pads into which speci-
men tubes could be placed. Each container was inserted in a cor-
rugated cardboard box to protect it during transport.
The triple packing system became widely adopted for sample
transportation, but the quality of the samples themselves re-
mained inconsistent. For example, no standardized swabbing
system was in use, and persistent problems were associated
with these specimens. Often the swab samples were dry and re-
quired rehydration with lysis buffer. The wooden shafts of
swabs were sometimes broken off, leaving the sharp, splintered
ends exposed when the specimen tube was opened; sometimes
the shafts were too long for the lid to be tted back on to the
tube, causing the lid to be loose and the tube contents to leak
inside the shipping container. One district repeatedly sent
swab samples in tubes wedged inside Vacutainer tubes, which
could not be retrieved. Two districts consistentlysent swab sam-
ples in bacterial agar transport medium. The CDC attempted to
provide swabs and tubes of virus transport medium to various
districts, but these did not always make it to the intended recip-
ients, and specimens prepared with these materials were only
rarely received by the laboratory.
It was sometimes difcult to link patients with their results;
multiple patients with identical names could be present in an
ETU simultaneously. On occasion, one identier was assigned
Figure 3. Personal protective equipment used within the hot laboratory
at the Centers for Disease Control and Prevention laboratory in Sierra Leone.
S6 JID Flint et al
at Stephen B. Thacker CDC Library on August 4, 2015http://jid.oxfordjournals.org/Downloaded from
Figure 4. A, Cumulative number of samples tested for Ebola virus (EBOV) RNA by the Centers for Disease Control and Prevention laboratory in Sierra
Leone, 22 August 2014 to 22 March 2015. B, Number of samples tested per day.
Ebola Field Laboratory JID S7
at Stephen B. Thacker CDC Library on August 4, 2015http://jid.oxfordjournals.org/Downloaded from
at a holding facility and a different identier was given at the
ETU. The use of unique patient identiers eventually solved
these issues, but this did not become standard practice until
many months into the outbreak.
Another challenge was the supply of electricity to the labora-
tory. The Bo laboratory received electrical power from a gener-
ator operated by MSF, which was reliable, for the most part.
Power failures did occasionally occur, and the ability of the Bio-
Rad real-time system to restart an assay at the point that it had
stopped, once the power supply resumed, proved extremely use-
ful; brief power outages did not affect the data quality. Another
useful feature of this instrument was that it does not require an
internal control uorophore, such as Rox, which simplied re-
action setup. To avoid the loss of power during RNA extrac-
tions, the instruments were generally powered by a portable
gasoline-powered generator (HondaEU2000i), though over-
heating or possibly the quality of the locally sourced fuel was
sometimes responsible for problems with these generators.
Laboratory Throughput
The laboratory received and processed specimens daily from 22
August 2014 through 22 March 2015, including the day of relo-
cation from Kenema to Bo, Thanksgiving, Christmas Day, and
New Years Day. More than 12 000 specimens were tested in
total (Figure 4A). (Note added in proof: at the time of manu-
script acceptance, more than 20 000 specimens had been test-
ed.) The mean number of samples tested per day was 63, with
the highest number tested being 178 (Figure 4B). Samples trans-
ported from far aeld sometimes arrived too late in the day for
processing and were tested the next morning, but on average,
71% of samples were processed on the day they arrived at the
laboratory, with the results distributed that evening. Samples
were held over for next-day processing for several reasons: to re-
duce fatigue on the team, for safety concerns due to low light in
the hot laboratory after dark, and, most importantly, if patients
would not be moved after dark and the laboratory results
would not be acted on until the next day. In all, 99.9% of samples
were tested either on the day of receipt orthe nextday. Full details
of the assay performance, the laboratory results, and their impact
on the outbreak response will be published elsewhere.
DISCUSSION
The role of the US CDC laboratory in Bo was to provide EBOV
diagnostics for 3 purposes: (1) testing blood drawn from pa-
tients in holding centers to guide their transfer to an ETU or
otherwise, (2) testing patients in the ETU to help with decisions
regarding their discharge, and (3) testing oral swab samples col-
lected from dead bodies to facilitate contact tracing and the im-
plementation of safe burial protocols. Although we believe that
the CDC laboratory was an important resource for the clini-
cians caring for EBOV patients across much of Sierra Leone,
improvements to the laboratory services were possible. Owing
to the excessive workloads, assays other than the EBOV RT-
PCR tests were not performed until early February 2015,
when daily sample numbers had dropped signicantly. At this
stage it proved possible to implement blood chemistry tests and
a rapid diagnostic assay for malaria.
Other tests that could have helped guide patient care include
testing for other infectious diseases, such as Lassa fever, shigello-
sis, and typhoid fever. Serological assays for anti-EBOV IgG and
IgM were occasionally requested by ETU clinicians. The CDC
laboratory did not have the capacity to perform these assays in
Sierra Leone. The IgM assay would have been useful to inform
contact tracing efforts. The IgG assay could have been used to as-
sess titers in the blood of EBOV disease survivors, before its trans-
fusion as an emergency experimental treatment. In these cases,
testing for human immunodeciency, hepatitis B, and hepatitis
C viruses would also have been useful. Such an application was
not a common occurrence, however.
Although the assays described here have been successfully
used under low-resource conditions, they are relatively expen-
sive and require some laboratory infrastructure and trained
staff. A simple, rapid, point-of-care diagnostic test that could
detect EBOV in blood from a nger prick would be invaluable
for testing in community settings. Such a test would need to be
sensitive, with a clear algorithm for retesting negative patients.
Several platforms are currently under evaluation, and will prob-
ably be deployed as a presumptive diagnostic test designed to
complement the existing RT-PCR assays.
In summary, we have described the organization and proce-
dures of the US CDC EBOV diagnostic laboratory located in Bo,
Sierra Leone. Visitors to this facility have commented on the
simplicity of the laboratory setup, and expressed surprise at
its throughput, diagnostic accuracy, and rapid turn-around in
support of the EBOV outbreak response in Sierra Leone. We
hope that this article aids the deployment and establishment
of other laboratories, to serve the patients of this EBOV out-
break and any future ones.
Notes
Acknowledgments. This article is dedicated to all those working to
combat the EBOV outbreak, especially the medical technicians and phlebot-
omists who have risked their lives drawing blood and delivering it to the lab-
oratory. We thank Issah French, Will Pooley, Mambu Momoh, Ian Crozier,
Kaci Hickox, Frederique Jacquerioz, Suzanne Donovan, Lewis Rubinson,
Darrio Gramuglia, Henry Kyobe, and Monia Sayah; your courage and devo-
tion to helping those aficted has awed and inspired us. We thank Tanya
Klimova for assistance with editing this manuscript. We thank the Sierra
Leone Ministry of Health and Sanitation, the World Health Organization
and the Global Outbreak Alert and Response Network for organizing the
deployment of Centers for Disease Control and Prevention (CDC) scientists
to Sierra Leone. We thank the nurses, physicians, and other employees of
Kenema Government Hospital and MSF Bo. We also thank the Internation-
al Rescue Committee for their support of Bo Government Hospital. The lab-
oratory scientist pictured in Figure 3provided explicit written consent for
use of the image.
S8 JID Flint et al
at Stephen B. Thacker CDC Library on August 4, 2015http://jid.oxfordjournals.org/Downloaded from
Disclaimer. The ndings and conclusions in this report are those of the
authors and do not necessarily represent the ofcial position of the CDC.
Potential conicts of interest. All authors: No reported conicts.
All authors have submitted the ICMJE Form for Disclosure of Potential
Conicts of Interest. Conicts that the editors consider relevant to the con-
tent of the manuscript have been disclosed.
References
1. Kuhn JH, Andersen KG, Baize S, et al. Nomenclature- and database-
compatible names for the two Ebola virus variants that emerged in
Guinea and the Democratic Republic of the Congo in 2014. Viruses
2014; 6:476099.
2. Sanchez A, Geisbert TW, Feldmann H. Filoviridae: Marburg and Ebola
viruses. In: Knipe DM, Howley PM, eds. Fields virology. Philadelphia,
PA: Lippincott Williams & Wilkins, 2006:140948.
3. Drosten C, Gottig S, Schilling S, et al. Rapid detection and quantica-
tion of RNA of Ebola and Marburg viruses, Lassa virus, Crimean-Congo
hemorrhagic fever virus, Rift Valley fever virus, dengue virus, and yel-
low fever virus by real-time reverse transcription-PCR. J Clin Microbiol
2002; 40:232330.
4. Leroy EM, Baize S, Lu CY, et al. Diagnosis of Ebola haemorrhagic fever
by RT-PCR in an epidemic setting. J Med Virol 2000; 60:4637.
5. Sanchez A, Ksiazek TG, Rollin PE, et al. Detection and molecular char-
acterization of Ebola viruses causing disease in human and nonhuman
primates. J Infect Dis 1999; 179(suppl 1):S1649.
6. Towner JS, Rollin PE, Bausch DG, et al. Rapid diagnosis of Ebola hem-
orrhagic fever by reverse transcription-PCR in an outbreak setting and
assessment of patient viral load as a predictor of outcome. J Virol 2004;
78:433041.
7. Ksiazek TG, Rollin PE, Williams AJ, et al. Clinical virology of Ebola
hemorrhagic fever (EHF): virus, virus antigen, and IgG and IgM anti-
body ndings among EHF patients in Kikwit, Democratic Republic of
the Congo, 1995. J Infect Dis 1999; 179(suppl 1):S17787.
8. Ksiazek TG, West CP, Rollin PE, Jahrling PB, Peters CJ. ELISA for the
detection of antibodies to Ebola viruses. J Infect Dis 1999; 179(suppl 1):
S1928.
9. Martines RB, Ng DL, Greer PW, Rollin PE, Zaki SR. Tissue and cellular
tropism, pathology and pathogenesis of Ebola and Marburg viruses.
J Pathol 2015; 235:15374.
10. Zaki SR, Shieh WJ, Greer PW, et al. A novel immunohistochemical
assay for the detection of Ebola virus in skin: implications for diagnosis,
spread, and surveillance of Ebola hemorrhagic fever. Commission de
Lutte contre les Epidemies a Kikwit. J Infect Dis 1999; 179(suppl 1):
S3647.
11. World Health Organization. Laboratory diagnosis of Ebola virus dis-
ease. http://apps.who.int/iris/bitstream/10665/134009/1/WHO_EVD_
GUIDANCE_LAB_14.1_eng.pdf?ua=1. Accessed 21 March 2015.
12. Centers for Disease Control and Prevention. Guidance for collection,
transport and submission of specimens for Ebola virus testing. http://
www.cdc.gov/vhf/ebola/healthcare-us/laboratories/specimens.html.
Accessed 21 March 2015.
Ebola Field Laboratory JID S9
at Stephen B. Thacker CDC Library on August 4, 2015http://jid.oxfordjournals.org/Downloaded from
... Utilization of the RCK for inactivation of blood and oral swab specimens and the Xpert Ebola Assay for EBOV testing (integrated nucleic acid extraction and qRT-PCR) required only a single laboratory room with two designated workspaces. In contrast, high-throughput VHF field laboratories such as the CDC Ebola Field Laboratory that was established in Sierra Leone during the 2014-2015 West African EVD Outbreak [13], typically require an outdoor workspace for specimen inactivation while donning full PPE and three separated indoor workspaces for: 1) the preparation of clean nucleic acid extraction reagents and qRT-PCR master mixes, 2) RNA extraction, and 3) the addition of RNA template to the qRT-PCR master mix. Electrical power at the field laboratory was only required when operating the GeneXpert Instrument, as the exhaust air pump for the RCK was battery-powered and the Xpert Ebola Assay cartridges did not require refrigeration. ...
... Larger EVD outbreak responses requiring an increased specimen throughput may want to consider using a 16-module GeneXpert Dx instrument, and additional RCKs and laboratory staff. Alternatively, if dependable electricity, highly trained laboratory staff, and sufficient space are available, standing-up a high-throughput field laboratory, such as the CDC Ebola Field Laboratory that was established in Sierra Leone during the 2014-2015 West African EVD Outbreak [13], should be considered. During this deployment of the field laboratory, we tapped into the hospital clinical laboratory's UPS to secure electricity for the GeneXpert Instrument. ...
Article
Full-text available
The Democratic Republic of the Congo (DRC) declared an Ebola virus disease (EVD) outbreak in North Kivu in August 2018. By June 2019, the outbreak had spread to 26 health zones in northeastern DRC, causing >2,000 reported cases and >1,000 deaths. On June 10, 2019, three members of a Congolese family with EVD-like symptoms traveled to western Uganda’s Kasese District to seek medical care. Shortly thereafter, the Viral Hemorrhagic Fever Surveillance and Laboratory Program (VHF program) at the Uganda Virus Research Institute (UVRI) confirmed that all three patients had EVD. The Ugandan Ministry of Health declared an outbreak of EVD in Uganda’s Kasese District, notified the World Health Organization, and initiated a rapid response to contain the outbreak. As part of this response, UVRI and the United States Centers for Disease Control and Prevention, with the support of Uganda’s Public Health Emergency Operations Center, the Kasese District Health Team, the Superintendent of Bwera General Hospital, the United States Department of Defense’s Makerere University Walter Reed Project, and the United States Mission to Kampala’s Global Health Security Technical Working Group, jointly established an Ebola Field Laboratory in Kasese District at Bwera General Hospital, proximal to an Ebola Treatment Unit (ETU). The laboratory consisted of a rapid containment kit for viral inactivation of patient specimens and a GeneXpert Instrument for performing Xpert Ebola assays. Laboratory staff tested 76 specimens from alert and suspect cases of EVD; the majority were admitted to the ETU (89.3%) and reported recent travel to the DRC (58.9%). Although no EVD cases were detected by the field laboratory, it played an important role in patient management and epidemiological surveillance by providing diagnostic results in <3 hours. The integration of the field laboratory into Uganda’s National VHF Program also enabled patient specimens to be referred to Entebbe for confirmatory EBOV testing and testing for other hemorrhagic fever viruses that circulate in Uganda.
... Real-time quantitative reverse-transcription polymerase chain reaction (qRT-PCR) is the current standard for confirming acute EVD. qRT-PCR inversely measures the quantity of viral nucleic acid by determining the number of cycles of RNA replication that have occurred when EBOV-specific RNA signal is detected, the cycle threshold (Ct) value, which can thus be considered a surrogate measure of viral load [12][13][14]. It is plausible that higher presumed viral load in a patient's acute specimen (lower Ct value) would be associated with increased rates of transmission, but this association has never been evaluated or reported. ...
... Geographic proximity to the healthcare facility where the index patient was evaluated and laboratory capacity were factors determining which laboratory performed specimen testing. Laboratories used different methods of qRT-PCR testing similar to those previously reported for the laboratory in Bo District [14] to detect EBOV RNA from blood (live patients) and oral swab (deceased patients) specimens. In addition to a qualitative test result (eg, positive or negative), laboratories reported Ct values for positive specimens, an inverse measure of EBOV RNA and a surrogate for viral load [12,13]. ...
Article
Background: Identifying risk factors for household transmission of Ebola virus (EBOV) is important to guide preventive measures during Ebola outbreaks. Methods: We enrolled all confirmed persons with EBOV disease who were the first case patient in a household from December 2014 to April 2015 in Freetown, Sierra Leone, and their household contacts. Index patients and contacts were interviewed, and contacts were followed up for 21 days to identify secondary cases. Epidemiologic data were linked to EBOV real-time reverse-transcription polymerase chain reaction cycle threshold (Ct) data from initial diagnostic specimens obtained from enrolled index case patients. Results: Ct data were available for 106 (71%) of 150 enrolled index patients. Of the Ct results, 85 (80%) were from blood specimens from live patients and 21 (20%) from oral swab specimens from deceased patients. The median Ct values for blood and swab specimens were 21.0 and 24.0, respectively (P = .007). In multivariable analysis, a Ct value from blood specimens in the lowest quintile was an independent predictor of both increased risk of household transmission (P = .009) and higher secondary attack rate among household contacts (P = .03), after adjustment for epidemiologic factors. Conclusions: Our findings suggest the potential to use Ct values from acute EBOV diagnostic specimens for index patients as an early predictor of high-risk households and high-risk groups of contacts to help prioritize EBOV disease investigation and control efforts.
... Laboratories that handle these pathogens, especially BSL-4, have very strict engineering controls such as separate or controlled ventilation, separate work areas for sample transfer, extraction and analysis, authorized access only, and special personal protective equipment such as full body and air supplied suits [109]. Attention must be given to containment and thorough disinfection techniques during sample processing since it is possible that pathogens will not be inactivated prior to molecular testing [110,111]. ...
Article
Recent studies evaluating the preanalytical factors that impact the outcome of nucleic-acid based methods for the confirmation of SARS-CoV-2 have illuminated the importance of identifying variables that promoted accurate testing, while using scarce resources efficiently. The majority of laboratory errors occur in the preanalytical phase. While there are many resources identifying and describing mechanisms for main laboratory testing on automated platforms, there are fewer comprehensive resources for understanding important preanalytical and environmental factors that affect accurate molecular diagnostic testing of infectious diseases. This review identifies evidence-based factors that have been documented to impact the outcome of nucleic acid-based molecular techniques for the diagnosis of infectious diseases.
... The CDC assumed responsibility for EBOV diagnostic testing at the KGH laboratory in late August 2014 [28]. The VHF laboratory at KGH continued to receive samples from the International Federation of Red Cross and Red Crescent Societies and other ETUs for Lassa fever and EVD diagnostic screening. ...
... As reported by other laboratories, 14 there were concerns regarding the handling of small vials in the glove box with the thick, oversized gloves required to maintain a safe barrier between the HRZ and LRZ, particularly because the glove ports blocked the technicians' field of view while they manipulated samples. The laboratory technicians also reported reduced capacity for dexterity and concerns regarding the contamination of the rims of the vials used in the GeneXpert analyzer. ...
Article
Full-text available
The capacity to rapidly distinguish Ebola virus disease from other infectious diseases and to monitor biochemistry and viremia levels is crucial to the clinical management of suspected Ebola virus disease cases. This article describes the design and practical considerations of a laboratory straddling the high- and low-risk zones of an Ebola treatment center to produce timely diagnostic and clinical results for informed case management of Ebola virus disease in real-life conditions. This innovation may be of relevance for actors requiring flexible laboratory implementation in contexts of high-communicability, high-lethality disease outbreaks.
Article
Infectious diseases continue to be the leading cause of morbidity and mortality, and a formidable obstacle to the development and well-being of people worldwide. Viruses account for more than half of infectious disease outbreaks that have plagued the world. The past century (1918/19-2019/20) has witnessed some of the worst viral disease outbreaks the world has recorded, with overwhelming impact especially in low- and middle-income countries (LMIC). The frequency of viral disease outbreak appears to be increasing. Generally, although infectious diseases have afflicted the world for centuries and humankind has had opportunities to examine the nature of their emergence and mode of spread, almost every new outbreak poses a formidable challenge to humankind, beating the existing pandemic preparedness systems, if any, and causing significant losses. These underscore inadequacy in our understanding of the dynamics and preparedness against viral disease outbreaks that lead to epidemics and pandemics. Despite these challenges, the past 100 years of increasing frequencies of viral disease outbreaks have engendered significant improvements in response to epidemics and pandemics, and offered lessons to inform preparedness. Hence, the increasing frequency of emergence of viral outbreaks and the challenges these outbreaks pose to humankind, call for the continued search for effective ways to tackle viral disease outbreaks in real time. Through a PRISMA-based approach, this systematic review examines the outbreak of viral diseases in retrospect to decipher the outbreak patterns, losses inflicted on humanity and highlights lessons these offer for meaningful preparation against future viral disease outbreaks and pandemics.
Article
Alternate care sites (ACSs) are temporary medical locations established in response to events that disrupt or limit the ability of established medical facilities to provide adequate care. As with established medical facilities, ACSs require careful consideration of infection prevention and control (IPC) practices to mitigate risk of nosocomial transmission and occupational exposure. We conducted a rapid systematic review of published literature from the date of inception of each database until the date the search was run (September 2021) on the IPC practices in ACSs. The practices described were categorized using the National Institute of Occupational Safety and Health hierarchy of controls framework, including elimination, substitution, engineering controls, administrative controls, and personal protective equipment. Of 313 articles identified, 55 were included. The majority (n=45, 81.8%) were case reports and described ACSs established in the context of infectious disease outbreaks (n=48, 87.3%), natural disasters (n=5, 9%), and military deployments (n=2, 3.6%). Implementation of engineering and/or administrative control practices predominated, with personal protective equipment emphasized in articles related to infectious disease outbreaks. These findings emphasize both a need for more high-quality research into the best practices for IPC in ACSs and how to incorporate the most effective strategies in these settings in response to future events.
Article
Background During the 2014-2016 Ebola outbreak in West Africa, the Sierra Leone Ministry of Health and Sanitation (MoHS), the US Centers for Disease Control and Prevention, and responding partners under the coordination of the National Ebola Response Center (NERC) and the MoHS's Emergency Operation Center (EOC) systematically recorded information from the 117 Call Center system and district alert phone lines, case investigations, laboratory sample testing, clinical management, and safe and dignified burial records. Since 2017, CDC assisted MoHS in building and managing the Sierra Leone Ebola Database (SLED) to consolidate these major data sources. The primary objectives of the project were helping families to identify the location of graves of their loved ones who died at the time of the Ebola epidemic through the SLED Family Reunification Program and creating a data source for epidemiological research. The objective of this paper is to describe the process of consolidating epidemic records into a useful and accessible data collection and to summarize data characteristics, strength, and limitations of this unique information source for public health research. Methods Because of the unprecedented conditions during the epidemic, most of the records collected from responding organizations required extensive processing before they could be used as a data source for research or the humanitarian purpose of locating burial sites. This process required understanding how the data were collected and used during the outbreak. To manage the complexity of processing the data obtained from various sources, the Sierra Leone Ebola Database (SLED) Team used an organizational strategy that allowed tracking of the data provenance and lifecycle. Results The SLED project brought raw data into one consolidated data collection. It provides researchers with secure and ethical access to the SLED data and serves as a basis for the research capacity building in Sierra Leone. The SLED Family Reunification Program allowed Sierra Leonean families to identify location of the graves of loved ones who died during the Ebola epidemic. Discussion The SLED project consolidated and utilized epidemic data recorded during the Sierra Leone Ebola Virus Disease outbreak that were collected and contributed to SLED by national and international organizations. This project has provided a foundation for developing a method of ethical and secure SLED data access while preserving the host nation's data ownership. SLED serves as a data source for the SLED Family Reunification Program and for epidemiological research. It presents an opportunity for building research capacity in Sierra Leone and provides a foundation for developing a relational database. Large outbreak data systems such as SLED provide a unique opportunity for researchers to improve responses to epidemics and indicate the need to include data management preparedness in the plans for emergency response.
Technical Report
Full-text available
developed the original conceptual framework for the project, secured funding, co-developed the research design, contributed to the analysis, provided feedback on the first draft and contributed to the writing of the final draft.
Article
Introduction: U.S. hospitals that admitted Ebola virus disease (EVD) patients mitigated risk by using point-or-care testing (POCT) for critical support in isolation units. Success proved unequivocally the need for POCT. Additionally, molecular diagnostics have been used to help stop new outbreaks, and even handheld diagnostic solutions are emerging. Areas covered: This update of “Molecular detection and point-of-care testing in Ebola virus disease and other threats” [Expert Reviews 2015;15(10):1249-1255], assesses the impact of EVD epidemics, documents insights from recent reviews, summarizes evolving POC molecular technologies, presents General Accountability Office (GAO) recommendations, identifies the role of POC Coordinators, and casts a vision for national POCT policies and guidelines. Factual updating comprised summarizing EVD outbreaks including 2017-18, analyzing reviews and evidence-based publications since the 2014-16 epidemic, and tabulating published technical and molecular diagnostics. New graphics illustrate POC error mitigation/risk reduction, a framework for national POCT policy and guidelines, modular adaptations for country-specific solutions, and a logic diagram for future progress embedding artificial intelligence. Expert Commentary. The U.S. is still not prepared for highly infectious diseases. Key is lack of community rapid response and resilience, which must be enhanced not via mechanisms distant, but instead by molecular diagnostics directly at critical points of need.
Article
Full-text available
In 2014, Ebola virus (EBOV) was identified as the etiological agent of a large and still expanding outbreak of Ebola virus disease (EVD) in West Africa and a much more confined EVD outbreak in Middle Africa. Epidemiological and evolutionary analyses confirmed that all cases of both outbreaks are connected to a single introduction each of EBOV into human populations and that both outbreaks are not directly connected. Coding-complete genomic sequence analyses of isolates revealed that the two outbreaks were caused by two novel EBOV variants, and initial clinical observations suggest that neither of them should be considered strains. Here we present consensus decisions on naming for both variants (West Africa: "Makona", Middle Africa: "Lomela") and provide database-compatible full, shortened, and abbreviated names that are in line with recently established filovirus sub-species nomenclatures.
Article
Full-text available
Ebola (EBO) viruses were detected in specimens obtained during the hemorrhagic fever outbreak among humans in Kikwit, Democratic Republic of the Congo (DRC), in 1995 (subtype Zaire) and during an outbreak of disease in cynomolgus macaques in Alice, Texas, and the Philippines in 1996 (subtype Reston). Reverse transcriptase—polymerase chain reaction assays were developed and proven effective for detecting viral RNA in body fluids and tissues of infected individuals. Little change was seen in the nucleotide or deduced amino acid sequences of the glycoprotein (GP) of these EBO virus subtypes compared with those of their original representatives (i.e., the 1976 Yambuku, DRC, EBO isolate [subtype Zaire] and the 1989 Philippines and Reston, Virginia, isolates [subtype Reston]). The nonstructural secreted GP (SGP), the primary product of the GP gene, was more highly conserved than the structural GP, indicating different functional roles or evolutionary constraints for these proteins. Significant amounts of SGP were detected in acutely infected humans.
Article
Full-text available
Laboratory diagnosis of Ebola hemorrhagic fever (EHF) is currently performed by virus isolation and serology and can be done only in a few high-containment laboratories worldwide. In 1995, during the EHF outbreak in the Democratic Republic of Congo, the possibility of using immunohistochemistry (IHC) testing of formalin-fixed postmortem skin specimens was investigated as an alternative diagnostic method for EHF. Fourteen of 19 cases of suspected EHF met the surveillance definition for EHF and were positive by IHC. IHC, serologic, and virus isolation results were concordant for all EHF and non-EHF cases. IHC and electron microscopic examination showed that endothelial cells, mononuclear phagocytes, and hepatocytes are main targets of infection, and IHC showed an association of cellular damage with viral infection. The finding of abundant viral antigens and particles in the skin of EHF patients suggests an epidemiologic role for contact transmission. IHC testing of formalin-fixed skin specimens is a safe, sensitive, and specific method for laboratory diagnosis of EHF and should be useful for EHF surveillance and prevention.
Article
Full-text available
Viral hemorrhagic fevers (VHFs) are acute infections with high case fatality rates. Important VHF agents are Ebola and Marburg viruses (MBGV/EBOV), Lassa virus (LASV), Crimean-Congo hemorrhagic fever virus (CCHFV), Rift Valley fever virus (RVFV), dengue virus (DENV), and yellow fever virus (YFV). VHFs are clinically difficult to diagnose and to distinguish; a rapid and reliable laboratory diagnosis is required in suspected cases. We have established six one-step, real-time reverse transcription-PCR assays for these pathogens based on the Superscript reverse transcriptase-Platinum Taq polymerase enzyme mixture. Novel primers and/or 5'-nuclease detection probes were designed for RVFV, DENV, YFV, and CCHFV by using the latest DNA database entries. PCR products were detected in real time on a LightCycler instrument by using 5'-nuclease technology (RVFV, DENV, and YFV) or SybrGreen dye intercalation (MBGV/EBOV, LASV, and CCHFV). The inhibitory effect of SybrGreen on reverse transcription was overcome by initial immobilization of the dye in the reaction capillaries. Universal cycling conditions for SybrGreen and 5'-nuclease probe detection were established. Thus, up to three assays could be performed in parallel, facilitating rapid testing for several pathogens. All assays were thoroughly optimized and validated in terms of analytical sensitivity by using in vitro-transcribed RNA. The >or=95% detection limits as determined by probit regression analysis ranged from 1,545 to 2,835 viral genome equivalents/ml of serum (8.6 to 16 RNA copies per assay). The suitability of the assays was exemplified by detection and quantification of viral RNA in serum samples of VHF patients.
Article
Full-text available
The largest outbreak on record of Ebola hemorrhagic fever (EHF) occurred in Uganda from August 2000 to January 2001. The outbreak was centered in the Gulu district of northern Uganda, with secondary transmission to other districts. After the initial diagnosis of Sudan ebolavirus by the National Institute for Virology in Johannesburg, South Africa, a temporary diagnostic laboratory was established within the Gulu district at St. Mary's Lacor Hospital. The laboratory used antigen capture and reverse transcription-PCR (RT-PCR) to diagnose Sudan ebolavirus infection in suspect patients. The RT-PCR and antigen-capture diagnostic assays proved very effective for detecting ebolavirus in patient serum, plasma, and whole blood. In samples collected very early in the course of infection, the RT-PCR assay could detect ebolavirus 24 to 48 h prior to detection by antigen capture. More than 1,000 blood samples were collected, with multiple samples obtained from many patients throughout the course of infection. Real-time quantitative RT-PCR was used to determine the viral load in multiple samples from patients with fatal and nonfatal cases, and these data were correlated with the disease outcome. RNA copy levels in patients who died averaged 2 log(10) higher than those in patients who survived. Using clinical material from multiple EHF patients, we sequenced the variable region of the glycoprotein. This Sudan ebolavirus strain was not derived from either the earlier Boniface (1976) or Maleo (1979) strain, but it shares a common ancestor with both. Furthermore, both sequence and epidemiologic data are consistent with the outbreak having originated from a single introduction into the human population.
Article
Ebolaviruses and marburgviruses include some of the most virulent and fatal pathogens known to humans. These viruses cause severe haemorrhagic fevers with case fatality rates ranging from 25% to 90%. The diagnosis of filovirus using formalin-fixed tissues from fatal cases poses a significant challenge. The most characteristic histopathological findings are seen in the liver: however findings overlap with many other viral and non-viral haemorrhagic diseases. The need to distinguish filovirus infections from other haemorrhagic fevers, particularly in areas with multiple endemic viral haemorrhagic agents, is of paramount importance. In this review we discuss the current state of knowledge of filovirus infections and their pathogenesis, including histopathological findings, epidemiology, modes of transmission, and filovirus entry and spread within host organisms. The pathogenesis of filovirus infections is complex and involves activation of the mononuclear phagocytic system with release of proinflammatory cytokines, chemokines and growth factors; endothelial dysfunction; alterations of the innate and adaptive immune systems; direct organ and endothelial damage from unrestricted viral replication late in infection; and coagulopathy. Although our understanding of the pathogenesis of filovirus infections has rapidly increased in the past few years, many questions remain unanswered.
Article
EIAs for IgG and IgM antibodies directed against Ebola (EBO) viral antigens have been developed and evaluated using sera of animals and humans surviving infection with EBO viruses. The IgM capture assay detected anti-EBO (subtype Reston) antibodies in the sera of 5 of 5 experimentally infected animals at the time they succumbed to lethal infections. IgM antibodies were also detected in the serum of a human who was infected with EBO (subtype Reston) during a postmortem examination of an infected monkey. The antibody was detectable as early as day 6 after infection in experimentally infected animals and persisted for <90 days. The IgG response was less rapid; however, it persisted for >400 days in 3 animals who survived infection, and it persisted for ∼10 years after infection in the sera of 2 humans. Although these data are limited by the number of sera available for verification, the IgM assay seems to have great promise as a diagnostic tool. Furthermore the long-term persistence of the IgG antibodies measured by this test strongly suggests that the ELISA will be useful in field investigations of EBO virus.
Article
Ebola hemorrhagic fever (EHF) patients treated at Kikwit General Hospital during the 1995 outbreak were tested for viral antigen, IgG and IgM antibody, and infectious virus. Viral antigen could be detected in virtually all patients during the acute phase of illness, while antibody was not always detectable before death. Virus was also isolated from patients during the course of their febrile illness, but attempts to quantify virus in Vero E6 cells by standard plaque assay were often unsuccessful. IgG and IgM antibody appeared at approximately the same time after disease onset (8–10 days), but IgM persisted for a much shorter period among the surviving convalescent patients. IgG antibody was detectable in surviving patients through about 2 years after onset, the latest time that samples were obtained. Detection of Ebola virus antigens or virus isolation appears to be the most reliable means of diagnosis for patients with suspected acute EHF, since patients with this often-fatal disease (80% mortality) may not develop detectable antibodies before death.
Article
This study reports the first field evaluation of a new diagnostic technique for Ebola virus disease with sensitivity and specificity. Ebola virus causes rare but fulminating outbreaks in Equatorial Africa. Rapid differentiation from other infections is critical for timely implementation of public health measures. Patients usually die before developing antibodies, necessitating rapid virus detection. A reverse transcriptase-polymerase chain reaction (RT-PCR) assay was developed, implemented and evaluated at Centre International de Recherches Médicales de Franceville (CIRMF) in Gabon, to detect Ebola viral RNA in peripheral blood mononuclear cells (PBMC). Twenty-six laboratory-confirmed patients during and 5 after the acute phase of Ebola haemorrhagic fever, 15 healthy controls and 20 febrile patients not infected with Ebola virus were studied. RT-PCR results were compared with ELISA antigen capture, and Ebola specific IgM and IgG antibody detection. Ebola virus RNA was amplified from 26/26 specimens from the acute phase, 3/5 during recovery, 0/20 febrile patients and 1/15 negative controls. Sensitivity of RT-PCR in identifying acute infection and early convalescence compared with antigen or IgM detection was 100% and 91% respectively, and specificity compared with antigen detection and IgM assay combined was 97%. Antigen capture detected only 83% of those identified by PCR, and IgM only 67%. Ebola virus RNA was detected in all 13 fatalities, only 5 of whom had IgM and none IgG. RT-PCR detected Ebola RNA in PBMC one to three weeks after disappearance of symptoms when antigen was undetectable. RT-PCR was the most sensitive method and able to detect virus from early acute disease throughout early recovery.