The variety of factors that contributed to the initial unde-
tectedspreadofEbolavirusdiseaseinWestAfricaduring
2013–2016andthedifcultycontrollingtheoutbreakonce
the etiology was identied highlight priorities for disease
prevention,detection,andresponse.Thesefactorsinclude
occurrenceinaregionrecoveringfromcivilinstabilityand
lackingexperiencewithEbolaresponse;inadequatesurveil-
lance,recognitionofsuspectedcases,andEboladiagnosis;
mobilepopulationsandextensiveurbantransmission;and
the community’s insufcient general understanding about
thedisease.Themagnitudeoftheoutbreakwasnotattrib-
utabletoasubstantialchangeofthevirus.Continuedefforts
duringtheoutbreakandinpreparationforfutureoutbreak
responseshouldinvolveidentifyingthereservoir,improving
in-countrydetectionandresponsecapacity,conductingsur-
vivorstudiesandsupportingsurvivors,engagingincultur-
allyappropriatepubliceducationandriskcommunication,
buildingproductiveinteragencyrelationships,andcontinu-
ingsupportforbasicresearch.
I
n 1976, the investigation of concurrent outbreaks of
a hemorrhagic fever syndrome (Ebola virus disease
[EVD]) in Zaire (currently Democratic Republic of Con-
go) and Sudan (currently Republic of South Sudan) (1,2)
led to isolation of 2 viruses now referred to as Ebola virus
(EBOV) and Sudan virus, respectively, and to identica-
tion of a newly recognized viral hemorrhagic fever genus,
Ebolavirus (family Filoviridae). Ebolaviruses now include
EBOV, Sudan virus, Reston virus, TForest virus and Bun-
dibugyo virus. The other genus in the family Filoviridae is
Marburgvirus, consisting of Marburg virus and Ravn virus
(termed marburgviruses; MBGV), both of which are asso-
ciated with severe disease (Marburg virus disease [MVD])
in humans (3,4). Before 2013, the largest Ebola outbreak
was associated with Sudan virus in Gulu, Uganda, in 2000
that caused 425 cases (224 fatal) (5). The largest EVD out-
break associated with EBOV (the same virus responsible
for the 2013–2016 outbreak in West Africa) was in Zaire
(1976) and caused 318 cases and an associated case-fatality
rate of 88% (2).
The EVD outbreak in Guinea, Liberia, and Sierra Leone
was unprecedented in its sheer magnitude and the emergence
of EBOV outside the Congo basin. The effect of the outbreak
is profound; as of March 27, 2016, a total of 28,646 EVD
cases and 11,323 deaths had been documented (6). Further-
more, this outbreak prompted an unparalleled international
response: 7 US agencies operated 9 laboratories, and 11
international agencies operated 13 laboratories performing
in-country diagnostic tests (Figure 1). The Centers for Dis-
ease Control and Prevention (CDC) supported ≈2,300 inter-
national deployments of ≈1,600 total personnel (both CDC
and non-CDC staff) (7); and thousands of personnel from
international aid agencies, e.g., World Health Organization,
Médecins Sans Frontières, International Rescue Committee,
International Finance Corporation, and Public Health Eng-
land provided in-country support.
The EVD outbreak was not restricted to the 3 heav-
ily affected West African countries; cases also occurred in
Senegal, Nigeria, and Mali. In addition, EBOV-infected
foreign aid workers were transported for treatment to Eu-
rope and the United States, and naturally imported cases
(United States, Italy, United Kingdom) and domestic trans-
mission (Spain, United States) were reported for the rst
time in several countries (6). The US EVD response includ-
ed establishment of EBOV testing in the US Laboratory
Response Network. As a result, 57 state, county, and local
public health laboratories in 44 states currently are quali-
ed to perform presumptive EBOV real-time quantitative
PCR (qPCR).
This EVD outbreak highlights globalization, interna-
tional social responsibility, and the importance of global
health security. As the response to the outbreak progresses,
the international research community must continue to ad-
dress questions of EBOV emergence, pathogenesis, and
transmission and advance therapeutic and vaccine develop-
ment. National and international organizations must criti-
cally assess the details of this outbreak and the correspond-
ing response to enable improved response and control of
emerging viral outbreaks.
Perspectives on West Africa
Ebola Virus Disease Outbreak,
2013–2016
Jessica R. Spengler, Elizabeth D. Ervin, Jonathan S. Towner, Pierre E. Rollin, Stuart T. Nichol
956 EmergingInfectiousDiseases•www.cdc.gov/eid•Vol.22,No.6,June2016
PERSPECTIVE
Authorafliation:CentersforDiseaseControlandPrevention,
Atlanta,Georgia,USA
DOI:http://dx.doi.org/10.32032/eid2206.160021
WestAfricaEbolaOutbreak
Why Here? Why Now?
We lack precise answers to these questions. A spillover
event is an exceedingly rare but high-consequence event
that is likely the most critical initiating factor for an out-
break. Features of the virus phylogenetic analyses and pu-
tative reservoir species, and what we know about prior ebo-
lavirus and MBGV spillover events, offer possible insight
into “Why here? Why now?” All currently recognized EB-
OVs appear to share a recent common ancestor 50 years
ago, probably because of a recent genetic bottleneck (8).
Current EBOV lineages, rst detected in northern Demo-
cratic Republic of Congo in 1976, appear to have spread
across the Congo basin during this relatively short period
and arrived to West Africa only in the past few years. Be-
fore the 2013–2016 outbreak, the only denitive evidence
of ebolaviruses or the diseases they cause in West Afri-
ca was 1 nonfatal human case associated with Taï Forest
virus, which caused illness and death in chimpanzees in
Côte d’Ivoire in 1994 (9).
How EBOV spread across the Congo basin and wheth-
er this spread involved movement through bat, nonhuman
primate, or other animal populations are unclear. EVD in
humans has been linked to preparing and eating nonhuman
primate (chimpanzee, gorilla, monkey) or duiker bushmeat
(10). Contact with bats also has been identied as a puta-
tive source of EBOV spillover (11). However, the role of
bats in virus maintenance and initiation of human disease
outbreaks remains unclear. Evidence of bats involvement in
the spillover event initiating the 2013–2016 outbreak is lim-
ited to anecdotal reports of interactions between bats and vil-
lagers in Guinea; no epidemiologic or genetic data associate
a putative reservoir species with the current outbreak (12).
Unlike MBGV, EBOV has yet to be isolated from bats, and
no direct evidence links bats to EBOV infection in humans.
EmergingInfectiousDiseases•www.cdc.gov/eid•Vol.22,No.6,June2016 957
Figure 1.Geographicdistribution
ofdiagnosticlaboratories
currentlyorpreviously
operationalinWestAfrica
duringthe2014–2015Ebola
virusresponse,asofDecember
9,2015.DataarefromWorld
HealthOrganizationEbolavirus
diseasesituationreports.
PERSPECTIVE
Regardless, epizootic spillover remains the most widely ac-
cepted theory for how the outbreak began.
Experimental infection studies with loviruses in-
dicate that the viral load in the carcass of an animal that
died of EVD would be high (13,14), but the viral load in
the carcass of a healthy reservoir species is probably much
lower (15,16). However, the virus inoculum required to
infect animal models of EVD by traditional experimental
routes is very low. Thus, viral shedding through excreta
or viral load in tissue (eaten or handled raw) of reservoir
species might provide sufcient inoculum to initiate virus
spillover. In spillover events involving MBGV, sequences
from human isolates were 99% identical to virus isolates
obtained directly from infected bats (17); thus, the EBOV
spillover event most likely involved little or no virus ad-
aptation. Most EBOV outbreaks appear to involve a single
initiating spillover event followed by human-to-human
transmission (18,19), whereas several MVD outbreaks
have been associated with multiple spillover events (4,17).
This dissimilarity might reect a difference in the nature
of human interactions with the different primary reservoir
species of EBOV and MBGV.
Why So Big?
Some early speculation about the differences in magnitude
between the 2013–2016 EVD outbreak and previous lo-
virus outbreaks was focused on the presence of a rapidly
mutating, highly transmissible or highly virulent EBOV
strain. Gire et al. reported a rapid accumulation of interhost
and intrahost genetic variation in 99 EBOV genomes from
78 patients in Sierra Leone (20). However, later analysis
of more EBOV full-length sequences indicated that the
overall virus nucleotide substitution rate was consistent
with rates observed in previous outbreaks in Central Africa
(8,21). Pathogenesis studies also support that the size of
the outbreak and characteristics of EVD in West Africa are
not related to change in the virus but instead appear to be a
result of factors extrinsic to the virus (2225).
Although differences in the outbreak strain do not ex-
plain the magnitude of the outbreak, the situation in these
West African countries in 2013 made them particularly
vulnerable to a large outbreak in the event of the arrival
of EBOV or spillover from wildlife. The West African
outbreak occurred in an extremely resource-poor area that
lacked basic infrastructure and was recovering from the
effects of decades of civil war. The consequences of civil
instability included the collapse of government institutions
and schools, disruption of traditional societal values and
structures, poor education standards, and struggling basic
healthcare infrastructures (2628). Unlike several other
African countries, Guinea, Liberia, and Sierra Leone had
no past experience in recognizing and managing lovirus
outbreaks, and the outbreak occurred in a region with very
high endemic levels of malaria that has a similar clinical
presentation to EVD. Although these countries had experi-
ence with Lassa hemorrhagic fever, that experience most
likely negatively affected the initial response: suspecting
Lassa might have delayed identifying EBOV and enabled
early EBOV transmission. Based on limited chains of hu-
man-to-human transmission Lassa virus appears to be less
transmissible and requires less stringent use of personal
protective equipment and containment to prevent health-
care worker infections.
Slow recognition of suspected cases, inability to accu-
rately diagnose disease, and absence of appropriate surveil-
lance for critical decision-making early in the outbreak se-
verely hampered interruption of EVD spread at key points
during the response. Distrust of government and outsiders
hindered response efforts, and the spread of conspiracy
theories among residents resulted in fear, superstition, and
secrecy (27). In contrast to prior lovirus outbreak where
treatment of cases and transmission occurred in major ur-
ban areas (e.g., Kinshasa, Zaire, in 1976; Nairobi, Kenya,
in 1980; Kinshasa in 1995; Johannesburg, South Africa, in
1995; Luanda, Angola, in 2005; Kampala, Uganda, 2007;
Kampala, Uganda, in 2014 [http://www.cdc.gov/vhf/ebola/
outbreaks/history/chronology.html; http://www.cdc.gov/
vhf/marburg/resources/outbreak-table.html]); the West Af-
rican outbreak was the rst to include multiple reintroduc-
tions to urban areas (such as Conakry) from human cases
and extensive urban transmission. Porous borders and high
population mobility within each country and into neighbor-
ing countries exacerbated widespread dissemination of dis-
ease from urban and rural transmission (27).
Future Priorities and Considerations
Identify the Reservoir
Predicting EBOV epizootics requires increased understand-
ing of virus ecology. Epidemiology, serologic data, and de-
tection of viral RNA support a role of bats and nonhuman
primates in EBOV maintenance and spillover transmission
from animal reservoirs to humans. However, EBOV has yet
to be isolated in nature from any bat species or nonhuman
primates. In contrast to EBOV, ecologic and experimen-
tal evidence conrms fruit bats (Rousettus aegyptiacus) as
a reservoir for MBGV, and MBGV spillover events from
bats to humans have been documented (4,17,29). Ecologic
investigations of R. aegyptiacus fruit bats showed seasonal
pulses of MBGV spillover events (30). MBGV has been
isolated from naturally infected R. aegyptiacus fruit bats 20
times (15,17,31), and virus replication and oral shedding in
the absence of clinical disease was observed in experimen-
tally infected R. aegyptiacus fruit bats (15).
Although EBOV exhibits ecologic patterns similar
to those of MBGV, conrming EBOV reservoir hosts by
958 EmergingInfectiousDiseases•www.cdc.gov/eid•Vol.22,No.6,June2016
WestAfricaEbolaOutbreak
virus isolation in nature remains elusive. One difculty
in obtaining EBOV isolates from bats, despite many at-
tempts, appears to be identifying and sampling the ap-
propriate bat species. Only 1, or a limited number of, bat
species most likely can serve as hosts for each of the lo-
virus species, a phenomenon also seen with rodentborne
hantaviruses and arenaviruses (32). In contrast to MBGV,
no detectable viremia develops in R. aegyptiacus fruit bats
experimentally infected with ebolaviruses (Sudan, Ebola,
Bundibugyo, Taï Forest, and Reston viruses), and viral
RNA detection was localized to the injection site (33),
suggesting that R. aegyptiacus fruit bats are not a compe-
tent reservoir species for EBOVs. MBGV ecologic studies
support the theory of 1, or a limited number of, host spe-
cies because infection was found consistently in R. aegyp-
tiacus fruit bats but not in Hipposideros spp. bats, despite
their close interaction (17).
Although bat species involved in EBOV maintenance
have yet to be discovered, limited detection of EBOV RNA
and EBOV antibodies has implicated some frugivorous and
insectivorous bat species distributed in areas of previous
outbreaks, including the little collared fruit bat (Myonycte-
ris torquata), hammer-headed bat (Hypsignathus monstro-
sus), Franquet’s epauletted fruit bat (Epomops franqueti),
straw-colored fruit bat (Eidolon helvum), and Angolan
free-tailed bat (Mops condylurus) (12,3436). Further in-
vestigation of these species as putative EBOV reservoirs
is warranted; identifying EBOV reservoir species would
enable predictive modeling based on distribution (Figure
2) and population dynamics (population size, reproduction,
proximity to human populations). Tracking fruit bat migra-
tions across their distribution ranges (Figure 2) including
riverine highways and conducting reservoir population sur-
veillance could identify high-risk disease foci before hu-
man population exposure, potentially mitigating another
spillover and outbreak. In the case of MBGV, understand-
ing the bat reservoir has led to risk reduction measures,
such as identifying seasons at high risk for spillover, re-
stricting access of miners or ecotourists to mines and caves
with circulating MBGV, and constructing a safe viewing
platform at a national park in Uganda.
Increase In-Country Surveillance, Diagnostic Capacity,
and Epidemiologic Support
Curbing disease spread requires rapid identication of the
initial human case or cluster after a spillover event to per-
mit patient isolation and timely contact tracing. The payoff
for investment in surveillance systems and diagnostic ca-
pacity to rapidly identify and respond to outbreaks is il-
lustrated by efforts in Uganda and Democratic Republic of
Congo, where the past several EVD and MVD outbreaks
were quickly identied and restricted to a few cases. Ef-
fective contact tracing to interrupt disease transmission is
personnel intensive and requires rapid organization and de-
ployment after notication of suspected cases. In all EVD
outbreaks to date, most transmission events involve close-
contact human-to-human transmission. Because resources
are often limited, case investigations should thoroughly
rule out known sources of EBOV transmission before in-
vestigating speculated or new (e.g., airborne, environmen-
tal, dogs, or asymptomatic human) sources that have never
been reported to be associated with disease in previous
lovirus outbreaks. Accomplishing the aforementioned
recommendations requires increased awareness at the com-
munity clinic level, improved quality of central laborato-
ries and capacity for specimen transport (rapid collection
and delivery, proper packaging, appropriate storage condi-
tions), and more epidemiologists throughout the region. Fi-
nally, laboratorians and epidemiologists must work closely
with well-trained clinical and infection control personnel
to effectively identify and isolate infectious persons (37).
Test Appropriate Samples and Interpret
Evidence-Based Data
Accurate diagnosis requires including EBOV in the differ-
ential diagnoses for febrile tropical illness and considering
the possibility of co-infection with other frequent tropical
diseases (e.g., malaria, typhoid). If appropriate diagnostic
samples are not collected, cases might be missed at critical
points in the response. Blood is the most sensitive diagnos-
tic specimen early in EVD; oral swabbing, although less
invasive, does not offer adequate sensitivity until late in
the course of disease, but it is a sensitive and appropriate
modality for testing postmortem specimens (13,14). Diag-
nostic data are critical in patient management and develop-
ment of clinical recommendations and discharge policies.
Interpretation of diagnostics should consider the sensitivity
and specicity of the test and specimen type and what the
test is detecting. For instance, qPCR is widely used, but a
common mistake in interpreting results is that RNA detec-
tion is synonymous with the presence of infectious virus or
viral shedding, which is not always the case (38). Although
detectable viral RNA might indicate shedding, it does not
equate with shedding, and this fact must be considered
when transmission risk is evaluated on the basis of qPCR.
Investigate Viral Persistence and Physical and
Psychological Sequelae in Survivors
The ≈17,000 survivors from the 2013–2016 EBOV outbreak
might face major medical and social challenges after recov-
ering from acute disease. Semen samples from EVD survi-
vors in Kikwit, Democratic Republic of Congo, were posi-
tive for EBOV by RT-PCR up to 101 days after illness onset,
and 1 sample obtained 82 days after disease onset yielded
infectious virus (19,39). In addition, MBGV was isolated
from semen of a convalescent patient and was the source of
EmergingInfectiousDiseases•www.cdc.gov/eid•Vol.22,No.6,June2016 959
PERSPECTIVE
infection of a contact (40; reference 41 in online Techni-
cal Appendix, http://wwwnc.cdc.gov/EID/article/22/6/16-
0021-Techapp1.pdf). Sexual transmission was also impli-
cated in EBOV transmission recently in Liberia (references
42,43 in online Technical Appendix). This outbreak con-
rmed that EBOV can persist in immune-privileged sites
and has highlighted the implications of these ndings.
EBOV has now been isolated from aqueous humor of the
eye (reference 44 in online Technical Appendix), semen
(17, reference 45 in online Technical Appendix), and cere-
brospinal uid (reference 46 in online Technical Appendix)
of patients in whom the initial viremia cleared. The details
and dynamics of EBOV shedding in body uids after con-
valescence remain unclear and need to be investigated fur-
ther, especially in uids with higher potential for involve-
ment in transmission events (e.g., semen and amniotic
uid) to clearly dene specimens and behaviors with trans-
mission risk.
Although understanding putative disease transmis-
sion from convalescent patients is essential to prevent
EVD, clinicians and public health professionals also must
investigate and address disease sequelae and social stigma
associated with EVD in affected populations. Convales-
cent EVD patients reported arthralgia and myalgia more
signicantly than control patients during the 1995 Kikwit
outbreak (39). In addition, 15% of the Kikwit survivors
interviewed reported ocular sequelae, including ocular
pain, photophobia, hyperlacrimation, and loss of visual
acuity; all 4 patients reporting ocular sequelae had uveitis
that responded to topical treatment (reference 47 in online
Technical Appendix). EVD survivors of the Bundibugyo
virus outbreak in Uganda in 2007 also had arthralgia and
960 EmergingInfectiousDiseases•www.cdc.gov/eid•Vol.22,No.6,June2016
Figure 2.RelationshipbetweenlocationofindexcaseinEbolavirus(Zaire ebolavirus)outbreaksandputativereservoirdistribution.
Ebolavirusoutbreaks(reddots)anddistributionofEidolon helvum,Mops condylurus,Myonycteris torquata,Epomops franqueti,and
Hypsignathus monstrosusbats(insets)areshown.DataarefromtheCentersforDiseaseControlandPreventionandtheInternational
UnionfortheConservationofNature.
WestAfricaEbolaOutbreak
ocular decits; hearing loss, neurologic abnormalities,
sleep disturbance, memory loss, and various other con-
stitutional symptoms. Chronic health problems also were
reported (references 48,49 in online Technical Appendix).
In addition to medical burdens associated with recov-
ery, survivors are concurrently dealing with considerable
psychological issues of fear, denial, and shame. In severe
instances, the social stigma associated with disease can be
profound, resulting in abandonment by family and friends
(reference 50 in online Technical Appendix).
Increase Public Education and Risk Communication
The response to an EVD outbreak requires rapid, effective,
widespread public education. In addition to increased poten-
tial for transmission, a lack of public education and knowl-
edge can contribute to panic, anxiety, and psychosocial
trauma; fear and distrust of treatment units and responders,
sometimes to the point of violence; and isolation, stigma-
tization, and community ostracism of survivors and family
members of patients (references 51,52 in online Technical
Appendix). Despite extensive communication efforts during
the current EVD outbreak, knowledge and understanding of
EVD symptoms remained low, and fear of ill and recovered
EVD patients and treatment units persists (reference 52 in
online Technical Appendix). To be effective, public educa-
tion must recognize community-specic risks and concerns.
This education must balance culturally appropriate messag-
ing in the context of scientically founded risk reduction
messages to minimize human exposure (references 5254
in online Technical Appendix). Risky behavior must be
identied, and messages about risky behavior, prevention
strategies, and feasible alternatives must be communicated.
Establishing in-country community partners should be in-
tegrated to health communication; these partners are often
most effective at providing behavioral health education and
overcoming language and cultural barriers (reference 54 in
online Technical Appendix).
Risk communication can prevent or greatly reduce
transmission. EBOV transmission occurs through close
contact with symptomatic EVD patients. Familial and so-
cial networks play a major role in transmission, particular-
ly through caregivers’ contact with infectious uids from
ill persons at home and in healthcare facilities and through
contact with deceased persons during funeral rites. Viral
transmission is relatively inefcient compared with other
highly infectious agents (reference 55 in online Technical
Appendix). An exception is the proposed contribution of
EBOV superspreaders (reference 56 in online Technical
Appendix): persons who infect disproportionally more
secondary contacts. For EBOV, superspreaders fall into
2 categories: biologic superspreaders, who shed more vi-
rus, and situational superspreaders, who solely because
of circumstances or behavior potentially expose more
persons, for example, persons who travel extensively,
have occupations that interact closely with many persons
(e.g., traditional healers), or deceased patients who had a
highly attended funeral. Although biologic superspread-
ers appear to occur in EVD outbreaks (references 57,58
in online Technical Appendix), situational superspreaders
more notably elicit transmission events, which successful
community education on EVD can greatly reduce.
Promote Productive Interagency Relationships
Overall outbreak prevention and response will benet
greatly from continuing efforts to develop relationships
with nongovernment organizations operating in the region
and encouragement of constructive reform of national and
international response agencies. We believe the very large
and complex nature of the outbreak made communication
within and among agencies exceptionally difcult during
the outbreak. Frequently, well-intended centralized deci-
sion-making did not translate into appropriate application
in the eld. During the outbreak, partnering among agen-
cies evolved in an effort to improve communication and
the outcome of collaborative efforts, but further improve-
ments are possible. Delegating roles among agencies in ac-
cordance with their strengths and abilities to acquire the
necessary resources for epidemiologic investigations, diag-
nostics, clinical care, media relations, public education, and
logistics might improve efciency.
Continue Support for Basic Research
Pathogenesis studies and development of diagnostic tests,
therapeutic drugs, and vaccines are the foundation of the
public heath response. The international scientic commu-
nity must continue to prioritize research on EBOV and viral
pathogens that have yet to manifest into large outbreaks
but have the fundamental characteristics to do so: viruses
causing high rates of illness and death that are capable of
person-to-person transmission and lack therapeutic drugs,
vaccines, and other interventions (e.g., Nipah virus and
Crimean-Congo hemorrhagic fever virus). The develop-
ment and study of new tools toward the end of an outbreak
is more likely to be hampered by a lack of patients, as we
observed with the EBOV vaccine trials and new diagnos-
tic test evaluations. Thus, future vital research projects
should be poised to deploy at the start of new outbreaks,
which will require prioritization and substantial regulatory
forethought and preparation. However, research projects
should not detract from outbreak response. The benets of
research investigations and fundamental response efforts
must be balanced appropriately. In response to the out-
break, EBOV researchers worked together in a remarkable
effort to advance research and address questions from the
eld in real time. Interagency collaborations and the open
communication of data should continue after the outbreak
EmergingInfectiousDiseases•www.cdc.gov/eid•Vol.22,No.6,June2016 961
PERSPECTIVE
to develop vaccines and therapeutic drugs and to address
key questions on EBOV and other high-consequence, high-
containment hemorrhagic fever viruses.
Conclusions
The large size and long duration of the West Africa EVD
outbreak and the resulting enormous national and interna-
tional response efforts yielded many lessons for improved
prevention and control efforts for emerging viral diseases.
Although the current outbreak comes to a close and other
health crises emerge in the news headlines, we must not
forget that many features of this tragic outbreak strongly re-
inforce the benet of continued investment in global health
security efforts.
Acknowledgments
We thank Tatyana Klimova for critical editing of the manuscript.
We also acknowledge and commend the international response
to the EVD outbreak in West Africa and thank the other
members of CDC’s Viral Special Pathogens Branch, Division of
High-Consequence Pathogens and Pathology, National Center
for Emerging and Zoonotic Infectious Diseases, for important
discussions and continued response efforts.
This work was supported in part by an appointment to the
Research Participation Program at CDC by the Oak Ridge
Institute for Science and Education through an interagency
agreement between the US Department of Energy and CDC (to
J.R.S.) and by the National Institutes of Health Loan Repayment
Award (to J.R.S.).
Dr. Spengler is a postdoctoral research fellow with the Viral
Special Pathogens Branch, National Center for Emerging and
Zoonotic Infectious Diseases, CDC. During the EVD outbreak
in West Africa, she assisted response efforts by providing
laboratory support domestically and by serving in the
Emergency Operations Center as a subject-matter expert for the
Laboratory Task Force and the Animal–Human Interface Team.
Her research interests include the pathogenesis, transmission,
and species barriers of viral hemorrhagic fevers.
References
1. World Health Organization. Ebola haemorrhagic fever in Sudan,
1976. Report of a WHO/International Study Team. Bull World
Health Organ. 1978;56:247–70.
2. World Health Organization. Ebola haemorrhagic fever in Zaire,
1976. Report of an international commission. Bull World Health
Organ. 1978;56:27193.
3. Johnson ED, Johnson BK, Silverstein D, Tukei P, Geisbert TW,
Sanchez AN, et al. Characterization of a new Marburg virus
isolated from a 1987 fatal case in Kenya. Arch Virol Suppl.
1996;11:101–14.
4. Bausch DG, Nichol ST, Muyembe-Tamfum JJ, Borchert M,
Rollin PE, Sleurs H, et al.; International Scientic and
Technical Committee for Marburg Hemorrhagic Fever Control in
the Democratic Republic of the Congo. Marburg hemorrhagic fever
associated with multiple genetic lineages of virus. N Engl J Med.
2006;355:909–19 http://dx.doi.org/10.1056/NEJMoa051465.
5. Towner JS, Rollin PE, Bausch DG, Sanchez A, Crary SM,
Vincent M, et al. Rapid diagnosis of Ebola hemorrhagic fever by
reverse transcription-PCR in an outbreak setting and assessment of
patient viral load as a predictor of outcome. J Virol. 2004;78:
4330–41 http://dx.doi.org/10.1128/JVI.78.8.4330-4341.2004.
6. World Health Organization. Ebola situation report. 2016 [cited
2016 Apr 25]. http://apps.who.int/ebola/ebola-situation-reports
7. Frieden TR, Damon IK. Ebola in West Africa—CDC’s role in
epidemic detection, control, and prevention. Emerg Infect Dis.
2015;21:1897–905 http://dx.doi.org/10.3201/eid2111.150949.
8. Carroll SA, Towner JS, Sealy TK, McMullan LK, Khristova ML,
Burt FJ, et al. Molecular evolution of viruses of the family
Filoviridae based on 97 whole-genome sequences. J Virol. 2013;
87:2608–16 http://dx.doi.org/10.1128/JVI.03118-12.
9. Formenty P, Hatz C, Le Guenno B, Stoll A, Rogenmoser P,
Widmer A. Human infection due to Ebola virus, subtype
Côte d’Ivoire: clinical and biologic presentation. J Infect Dis.
1999;179(Suppl 1):S48–53 http://dx.doi.org/10.1086/514285.
10. Leroy EM, Rouquet P, Formenty P, Souquière S, Kilbourne A,
Froment J-M, et al. Multiple Ebola virus transmission events and
rapid decline of central African wildlife. Science. 2004;303:387–90
http://dx.doi.org/10.1126/science.1092528.
11. Leroy EM, Epelboin A, Mondonge V, Pourrut X, Gonzalez J-P,
Muyembe-Tamfum J-J, et al. Human Ebola outbreak resulting
from direct exposure to fruit bats in Luebo, Democratic Republic
of Congo, 2007. Vector Borne Zoonotic Dis. 2009;9:723–8
http://dx.doi.org/10.1089/vbz.2008.0167.
12. Marí Saéz A, Weiss S, Nowak K, Lapeyre V, Zimmermann F,
Düx A, et al. Investigating the zoonotic origin of the West African
Ebola epidemic. EMBO Mol Med. 2014;7:17–23 http://dx.doi.org/
10.15252/emmm.201404792.
13. Prescott J, Bushmaker T, Fischer R, Miazgowicz K, Judson S,
Munster VJ. Postmortem stability of Ebola virus. Emerg Infect Dis.
2015;21:856–9 http://dx.doi.org/10.3201/eid2105.150041.
14. Spengler JR, Chakrabarti AK, Coleman-McCray JD, Martin BE,
Nichol ST, Spiropoulou CF, et al. Utility of oral swab sampling
for Ebola virus detection in guinea pig model. Emerg Infect Dis.
2015;21:1816–9 http://dx.doi.org/10.3201/eid2110.150840.
15. Amman BR, Jones MEB, Sealy TK, Uebelhoer LS, Schuh AJ,
Bird BH, et al. Oral shedding of Marburg virus in experimentally
infected Egyptian fruit bats (Rousettus aegyptiacus). J Wildl Dis.
2015;51:113–24 http://dx.doi.org/10.7589/2014-08-198.
16. Paweska JT, Jansen van Vuren P, Masumu J, Leman PA,
Grobbelaar AA, Birkhead M, et al. Virological and serological
ndings in Rousettus aegyptiacus experimentally inoculated with
vero cells-adapted hogan strain of Marburg virus. PLoS One.
2012;7:e45479 http://dx.doi.org/10.1371/journal.pone.0045479.
17. Towner JS, Amman BR, Sealy TK, Carroll SA, Comer JA,
Kemp A, et al. Isolation of genetically diverse Marburg viruses
from Egyptian fruit bats. PLoS Pathog. 2009;5:e1000536
http://dx.doi.org/10.1371/journal.ppat.1000536.
18. Ladner JT, Wiley MR, Mate S, Dudas G, Prieto K, Lovett S,
et al. Evolution and spread of Ebola virus in Liberia, 2014–2015.
Cell Host Microbe. 2015;18:659–69 http://dx.doi.org/10.1016/
j.chom.2015.11.008.
19. Rodriguez LL, De Roo A, Guimard Y, Trappier SG, Sanchez A,
Bressler D, et al. Persistence and genetic stability of Ebola virus
during the outbreak in Kikwit, Democratic Republic of the Congo,
1995. J Infect Dis. 1999;179(Suppl 1):S170–6 http://dx.doi.org/
10.1086/514291.
20. Gire SK, Goba A, Andersen KG, Sealfon RS, Park DJ, Kanneh L,
et al. Genomic surveillance elucidates Ebola virus origin and
transmission during the 2014 outbreak. Science. 2014;345:1369–72
http://dx.doi.org/10.1126/science.1259657.
962 EmergingInfectiousDiseases•www.cdc.gov/eid•Vol.22,No.6,June2016
WestAfricaEbolaOutbreak
21. Hoenen T, Safronetz D, Groseth A, Wollenberg KR, Koita OA,
Diarra B, et al. Virology. Mutation rate and genotype variation of
Ebola virus from Mali case sequences. Science. 2015;348:117–9
http://dx.doi.org/10.1126/science.aaa5646.
22. Albariño CG, Wiggleton Guerrero L, Lo MK, Nichol ST,
Towner JS. Development of a reverse genetics system to generate
a recombinant Ebola virus Makona expressing a green uorescent
protein. Virology. 2015;484:259–64 http://dx.doi.org/10.1016/
j.virol.2015.06.013.
23. Dunham EC, Banadyga L, Groseth A, Chiramel AI, Best SM,
Ebihara H, et al. Assessing the contribution of interferon
antagonism to the virulence of West African Ebola viruses.
Nat Commun. 2015;6:8000 http://dx.doi.org/10.1038/ncomms9000.
24. Marzi A, Feldmann F, Hanley PW, Scott DP, Günther S,
Feldmann H. Delayed disease progression in cynomolgus
macaques infected with Ebola virus Makona strain. Emerg Infect
Dis. 2015;21:1777–83 http://dx.doi.org/10.3201/eid2110.150259.
25. Bird BH, Spengler JR, Chakrabarti AK, Khristova ML,
Sealy TK, Coleman-McCray JD, et al. Humanized mouse model of
Ebola virus disease mimics immune responses in human disease.
J Infect Dis. 2016;213:703–11 http://dx.doi.org/10.1093/infdis/
jiv538.
26. McPake B, Witter S, Ssali S, Wurie H, Namakula J, Ssengooba F.
Ebola in the context of conict affected states and health systems:
case studies of Northern Uganda and Sierra Leone. Con Health.
2015;9:23 http://dx.doi.org/10.1186/s13031-015-0052-7.
27. World Health Organization. One year into the Ebola epidemic:
a deadly, tenacious and unforgiving virus [cited 2016 Jan 4].
http://www.who.int/csr/disease/ebola/one-year-report/
introduction/en/
28. Bausch DG, Schwarz L. Outbreak of ebola virus disease in Guinea:
where ecology meets economy. PLoS Negl Trop Dis. 2014;8:e3056
http://dx.doi.org/10.1371/journal.pntd.0003056.
29. Adjemian J, Farnon EC, Tschioko F, Wamala JF, Byaruhanga E,
Bwire GS, et al. Outbreak of Marburg hemorrhagic fever among
miners in Kamwenge and Ibanda Districts, Uganda, 2007.
J Infect Dis. 2011;204(Suppl 3):S796–9 http://dx.doi.org/10.1093/
infdis/jir312.
30. Amman BR, Carroll SA, Reed ZD, Sealy TK, Balinandi S,
Swanepoel R, et al. Seasonal pulses of Marburg virus circulation
in juvenile Rousettus aegyptiacus bats coincide with periods of
increased risk of human infection. PLoS Pathog. 2012;8:e1002877
http://dx.doi.org/10.1371/journal.ppat.1002877.
31. Amman BR, Nyakarahuka L, McElroy AK, Dodd KA, Sealy TK,
Schuh AJ, et al. Marburgvirus resurgence in Kitaka Mine bat
population after extermination attempts, Uganda. Emerg Infect Dis.
2014;20:1761–4 http://dx.doi.org/10.3201/eid2010.140696.
32. Mills JN, Childs JE. Ecologic studies of rodent reservoirs: their
relevance for human health. Emerg Infect Dis. 1998;4:529–37
http://dx.doi.org/10.3201/eid0404.980403.
33. Jones ME, Schuh AJ, Amman BR, Sealy TK, Zaki SR, Nichol ST,
et al. Experimental inoculation of Egyptian rousette bats (Rousettus
aegyptiacus) with viruses of the Ebolavirus and Marburgvirus
genera. Viruses. 2015;7:3420–42 http://dx.doi.org/10.3390/v7072779.
34. Leroy EM, Kumulungui B, Pourrut X, Rouquet P, Hassanin A,
Yaba P, et al. Fruit bats as reservoirs of Ebola virus. Nature.
2005;438:575–6 http://dx.doi.org/10.1038/438575a.
35. Pourrut X, Délicat A, Rollin PE, Ksiazek TG, Gonzalez JP,
Leroy EM. Spatial and temporal patterns of Zaire ebolavirus anti-
body prevalence in the possible reservoir bat species. J Infect Dis.
2007;196(Suppl 2):S176–83 http://dx.doi.org/10.1086/520541.
36. Ogawa H, Miyamoto H, Nakayama E, Yoshida R, Nakamura I,
Sawa H, et al. Seroepidemiological prevalence of multiple species
of loviruses in fruit bats (Eidolon helvum) migrating in Africa.
J Infect Dis. 2015;212(Suppl 2):S101–8 http://dx.doi.org/10.1093/
infdis/jiv063.
37. Chowell D, Castillo-Chavez C, Krishna S, Qiu X, Anderson KS.
Modelling the effect of early detection of Ebola. Lancet Infect Dis.
2015;15:148–9 http://dx.doi.org/10.1016/S1473-3099(14)71084-9.
38. Spengler JR, McElroy AK, Harmon JR, Ströher U, Nichol ST,
Spiropoulou CF. Relationship between Ebola virus real-time
quantitative polymerase chain reaction–based threshold cycle
value and virus isolation from human plasma. J Infect Dis.
2015;212(Suppl 2):S346–9 http://dx.doi.org/10.1093/infdis/
jiv187.
39. Rowe AK, Bertolli J, Khan AS, Mukunu R, Muyembe-Tamfum JJ,
Bressler D, et al. Clinical, virologic, and immunologic follow-
up of convalescent Ebola hemorrhagic fever patients and their
household contacts, Kikwit, Democratic Republic of the Congo.
Commission de Lutte contre les Epidémies à Kikwit. J Infect Dis.
1999;179(Suppl 1):S28–35 http://dx.doi.org/10.1086/514318.
40. Martini GA, Schmidt HA. Spermatogenic transmission of the
“Marburg virus”. (Causes of “Marburg simian disease”) [in
German]. Klin Wochenschr. 1968;46:398–400 http://dx.doi.
org/10.1007/BF01734141.
Address for correspondence: Stuart T. Nichol, Centers for Disease
Control and Prevention, 1600 Clifton Rd NE, Mailstop G14, Atlanta, GA
30329-4027, USA; email: [email protected]
EmergingInfectiousDiseases•www.cdc.gov/eid•Vol.22,No.6,June2016 963
The incidence of severe Haemophilus influenza infections,
such as sepsis and meningitis, has declined substantially
since the introduction of the H. influenzae serotype b
vaccine. However, the H. influenzae type b vaccine
fails to protect against nontypeable H. influenzae
strains, which have become increasingly frequent causes of
invasive disease, especially among children and the elderly.
Visit our website to listen:
hp://www2c.cdc.gov/podcasts/player.asp?f=8639125
EID Podcast: Invasive Disease Caused by
Nontypeable Haemophilus Influenzae