SlideShare a Scribd company logo
Examen écrit
Université de Genève
Faculté de Médecine
Ebola, an update
Emilie BRANCHE
Directeur de thèse: Prof. Francesco NEGRO
Co-directrice de thèse: Dr. Sophie CLÉMENT
Examinateurs: Prof. Laurent KAISER
Dr. Glauciá PARANHOS-BACCALA
2015
2
Abstract
The current outbreak of Ebola principally affecting Guinea, Sierra Leone and Liberia has
caused at least 24 000 infections and over 9 000 deaths as of March 2015. The "imported"
cases in Spain, United Kingdom and United States of America lead to an important surge of
media, medical and research interest. This review summarizes the current knowledge about
Ebola virus including the epidemiology, the pathogenesis as well as the treatments both
currently available and in development. We will see that Ebola pathogenesis is definitely
complex and that both coagulation and immune response play a crucial role in its etiology.
I - Introduction
1- Classification
Ebola virus belongs to the Ebolavirus genus in the Filoviridae family in the Mononegavirales
order. Filoviridae family includes Marburgvirus with Marburgvirus, Cuevavirus with Lloviu
cuevavirus and Ebolavirus. Since the original description in 1976 of the Zaire Ebolavirus , four
species have emerged, Bundibugyo, Cote d'Ivoire or Tai Forest, Reston and Sudan ebolavirus
[1] (Figure 1).
Figure 1 : Taxonomy of the Filoviridae family adapted from Kuhn JH et al, 2010 [2]
3
All these species are pathogenic for humans, except Reston ebolavirus, which is pathogenic
for non-human primates. These viruses have been sequenced and their molecular evolution
described [3]. The current circulating Ebolavirus (EBOV) shares 97% homology with Zaire
Ebolavirus [4]. This review will mainly focus on EBOV.
2 - Epidemiology
The first Ebola cases appeared in 1976 in Sudan [5] and in Zaire [6]. In southern Sudan, 284
cases appeared causing 151 deaths (53% of death) in 6 months (June to November). In
northern Zaire (near the Ebola river giving the name to this virus), 318 cases were described
leading to 280 deaths (88% of death) in 2 months (September and October).
Since these original cases, approximately 20 additional outbreaks occurred between 1976
and 2013, causing nearly 2500 deaths in the Democratic Republic of Congo, Sudan, Gabon,
Ivory Coast, Uganda and Zaire [7, 8]. Since December 2013, a new outbreak is occurring in
several African countries (Mali, Senegal, Nigeria, Guinea, Sierra Leone and Liberia). One case
has been described in United Kingdom, another in Spain and 4 cases in the United States of
America. This outbreak caused 24 282 infections so far with a mortality rate of 40% [9]
(Figure 2 and Figure S1).
Figure 2 : 8 March 2015: situation of EBOV outbreak.
4
a - Reservoir
Although the primary animal host for EBOV is still unclear, fruit bats seem to be its reservoir.
After several outbreaks in Gabon and Zaire between 2001 and 2005 that devastated local
gorilla and chimpanzee populations, a team of researchers captured 1030 animals including
bats, birds and small terrestrial vertebrates close to infected gorilla and chimpanzee
carcasses. They could detect immunoglobulin G (IgG) specific for EBOV virus in sera from
three different bat species, Hypsignathus monstrosus, Epomops franqueti and Myonycteris
torquata. Moreover, viral genome was detected in the same bat populations in organs
known to be the principal targets of EBOV, namely the liver and the spleen [10]. However,
many questions regarding the mechanism by which bats are infected and transmit the
viruses remain unanswered. Importantly, the transmission is not only due to direct contact
between human or non-human primates and living or dead bats. Indeed the majority of new
infections during outbreaks were due to human-human contacts through blood, secretions
or body fluids including sweat, saliva and tears [11].
b - Clinical characteristics
During the early stages, EBOV infection triggers several symptoms such as fever, severe
headache, muscle pain, intense weakness, fatigue, diarrhea, vomiting and abdominal
(stomach) pain. During the intermediate or advanced stages, inflammatory factors-induced
vasodilatation results in both internal and external hemorrhages (bleeding or bruising). In
addition to coagulation system disorders, the infection of kidney and liver leads to organ
dysfunctions. Body injury and viral spread in blood circulation and organs lead to a vicious
downward spiral. If viral spread cannot be controlled, patients may succumb to organ failure
or secondary bacterial infection. Hemorrhage observed during EBOV disease is due to
disseminated intravascular coagulation (DIC) development. This pathology is characterized
by a widespread activation of clotting cascade resulting in blood clots formation in blood
vessels, impairing the tissue blood flow and leading to ischemia and organ damages. In
addition, this blood clot formation exhausts the coagulation factors, preventing normal
coagulation and leading to hemorrhages. The mechanisms leading to DIC will be further
detailed below (see chapter III.2). Furthermore, there is a weak inflammatory response
coupled with a significant lymphoid cell apoptosis leading to lymphopenia, which seems to
be a marker of poor prognosis. These symptoms may appear anywhere from 2 to 21 days
after EBOV exposure, but the average is between 8-10 days. In most cases, the cause of
death is mainly due to organ failure (such as liver or spleen) rather than to hemorrhagic
fever. A major complication for the EBOV diagnostic is that symptoms observed during the
early stages of infection are non-specific and difficult to distinguish from other endemic
diseases, such as Lassa fever, malaria, cholera or typhoid fever.
5
II - The virus
1 - Morphology and genome organization
Viral particles have a filamentous morphology. The origin of the name of Filoviridae family
comes from the Latin word filum referring to this particular morphology (Figure 3a). EBOV is
an enveloped, single strand, non-segmented, negative sense RNA virus. The 19kb viral
genome contains seven genes separated by regulatory regions composed of the 3'
nontranslated region (NTR), highly conserved transcription stop and start signals and the
5'NTR. The conserved transcription stop and start signal either overlap or are separated by
intergenic regions (IGR) [12]. Even if the viral genome contains only seven genes, more
proteins are produced through cotranscriptional editing of the GP (glycoprotein) gene [13].
Encoded proteins are nucleoprotein (NP), polymerase cofactor VP35, matrix protein VP40,
glycoprotein (GP), transcriptional activator VP30, second matrix protein VP24 and RNA-
dependent-RNA polymerase (L) proteins. In addition, through RNA editing EBOV is able to
express two truncated secreted proteins, glycoprotein (sGP) and small glycoprotein (ssGP)
[13] (Figure 3b). The viral RNA is encapsidated by NP and associated to VP35, VP30 and L to
form the ribonucleoprotein (RNP) complex. The RNP is surrounded by a matrix structure,
containing the matrix proteins VP40 and VP24, and finally by a host cell-derived membrane
in which the surface glycoprotein GP is embedded [14]. GP self-associates as a trimer, linked
by a single disulfide bond to form spikes at the virion surface [15] (Figure 3c). In addition to
their structural function, these proteins play several roles notably in immune system evasion
as described below (see the chapter V)
Figure 3 : EBOV morphology observed by electron microscopy (a) (CDC, 2005) and schematic organization of genome (b)
[14] and virion (c) [16]
6
2 - EBOV protein functions
a - RNP complex
NP, VP35, VP30 and L proteins play a fundamental role in RNP complex formation and in viral
transcription and replication.
NP is a 739 amino-acids (aa) protein encoded by the first gene located at the 3' region of the
genome. It plays a central role in virus replication, NP together with VP24 and VP35 are
necessary and sufficient for the formation of nucleocapsids that are morphologically
indistinguishable from those from EBOV infected cells [17].
VP35 protein is composed of 321 aa (35kDa). In addition to its role in nucleocapsid formation
by creating a link between L and N, VP35 is also a cofactor of the RNA-dependent RNA
polymerase complex. It plays an important role in antiviral and IFN response inhibition
detailed later (see chapter V.2).
VP30 (288aa, 32kDa) interacts also with NP in the RNP complex. VP30 is a transciptional
activator. VP30 can switch from a phosphorylated inactive state to an active state, through
dephosphorylation by the cellular protein phosphatase 1 (PP1). This regulation maintains the
balance between transcription and replication, as VP30 activity is required for the
transcription initiation [18]. When VP30 is active, transcription and protein synthesis occur,
while when it is inactive, viral replication takes place.
The L gene encodes a large protein of 2212 aa (252kDa), highly conserved across Ebola
species. This RNA-dependent-RNA polymerase (similar to the other polymerases of negative
single stranded RNA virus) is responsible for the viral transcription as well as for the RNA
replication. Moreover, it regulates the GP editing leading to the generation of sGP and ssGP.
b - Matrix proteins: VP40 and VP24
VP40 is composed of 326 aa (35kDa) and is the most conserved and the most abundant
protein in the virion. It is not clear whether the majority of VP40 in the cytoplasm or
premembrane zone is monomeric [19-22] or dimeric [23] or both [24]. This VP40 form was
found to be critical for both the transport of the nucleocapsid to the cell surface and for its
incorporation into virions [23]. Nevertheless, this monomeric or dimeric conformation can
be switched into either octameric or hexameric structures that have distinct functions.
Octamer formation is critically dependent on RNA binding [25], as no octamer can be
observed in the absence of RNA [26], suggesting that they may play an important role in
EBOV transcription and replication [20]. Hexamers are believed to be induced by the VP40
binding to plasma membrane [19, 27] and may be implicated in the initiation of virus
assembly, binding and budding via their interaction with the cytoplasmic tails of viral GP
and/or the RNP complex [28, 29]. Hexameric VP40 induces host cell membrane curvature
7
needed for viral egress [24, 30]. This matrix protein plays a central role in the formation of
the filamentous structure of EBOV virions [23, 29]. However, how VP40 induces the
formation of the particular filamentous morphology of the particle is mostly unknown. In
addition, a soluble secreted form of VP40 was observed during EBOV infection in vitro and
was also found in the serum of virus-infected animals albeit in low amounts [31]. The role of
this soluble form of VP40 as well as the mechanism by which it is released are unknown.
Nevertheless, the early appearance of anti-VP40 antibodies in EBOV infected patients could
be explained by the presence of this secreted VP40 [32, 33]. These observations suggest that
soluble VP40 may play a role in EBOV pathogenicity.
VP24, composed of 251 aa (28kDa), plays a structural role of matrix but has also a function
during EBOV life cycle. In contrast to what has been reported in previous studies, Watt A et
al (2014) demonstrated that VP24 has only a very modest influence on genome replication
and transcription. Nevertheless, it plays an important role in particle infectivity due to its
function in nucleocapsid assembly and more specifically in RNA incorporation into viral
particles [34]. Like VP35, VP24 interferes with IFN response (see below in chapter V.2).
c - GP
The GP gene of EBOV contains an editing site allowing the translation of three differents
proteins (Figure 4). The first isoform is a structural protein, translated into a glycoprotein
precursor (GP0) further cleaved by a cellular proprotein convertase furin [35]. This produces
a surface subunit GP1 and a transmembrane subunit GP2 that are able to form a
heterotrimer. GP plays a role in virion attachment and fusion but this process remains poorly
understood. GP1 contains an excessively O-linked glycosylated mucin-like region (MLR) at C-
terminal, a heavily N-linked glycosylated glycan cap domain (GCD) and a receptor binding
domain (RBD) which mediate the binding to a variety of host cell surface factors including T-
cell immunoglobulin and mucin domain 1 (Tim-1) [36]. MLR is required for neither the viral
entry nor the cellular tropism [15], as MLR-deleted GP is able to mediate viral attachment
and entry, but it may influence the EBOV capacity to escape the immune system [37]. GP2
with the fusion peptide is required for the virus-host membrane fusion. In addition to this
transmembrane GP form, several soluble GPs have been described. A trimeric soluble GP,
called shed GP, is produced by the release of virion-attached GP byTNF-α-converting enzyme
(TACE) through a cleavage site proximal to the transmembrane anchor. Moreover, GP gene
encodes two non-structural forms of GP that are soluble and secreted in important quantity
by infected cells. The soluble GP (sGP) is homodimeric whereas the small soluble GP (ssGP) is
monomeric. During EBOV infection, the ratio between sGP and GP transcripts is
approximately 75% / 20% and ssGP represents 5% of GP transcripts [38]. These secreted GPs
are easily detectable in the blood of infected patients [39] and play several roles in both
cytoxicity induced by EBOV and immune evasion as detailed later (see chapter III.3 and
8
V.1.b). In addition, a study demonstrated that sGP can substitute GP1 to form sGP-GP2
complex, suggesting a role for sGP as a structural protein [40].
Figure 4 : Processing of EBOV glycoproteins from Cook et al, 2013 [41]
3 - Viral life cycle
EBOV life cycle is similar to life cycles of other viruses with negative single strand RNA
(Figure 5). After GP binding to attachment factors (including DC-SIGN, L-SIGN) [42] and entry
receptors, such as Tim-1 [43, 44], whole virions are internalized via macropinocytosis and
trafficked to the endosomal compartment [45, 46]. GP1 is then cleaved by the endosomal
cysteine proteases cathepsin B (CatB) and L (CatL) that remove the hyper-glycosylated
region, which exposes the RBD in order to bind the Niemann-Pick C1 (NPC1) cholesterol
transporter. GP1-NPC1 interaction leads to conformation change of trimeric GPs and allows
the insertion of three fusion peptides located at the N-terminal region of GP2 in endosomal
membrane. This step is essential for the fusion process, allowing viral genome release into
the cytoplasm [47, 48]. The released viral RNA is then first transcribed. Due to the presence
of transcription stop and start signal in the regulatory region between each gene, the
negative-strand RNA genome is transcribed by the L polymerase into seven monocistronic
mRNAs. These mRNAs are capped and polyadenylated. It is believed that for EBOV, such as
for all negative RNA viruses, the polymerase accesses to the viral genes via a single
9
polymerase binding site at the 3' end. Once bound the viral polymerase progresses along the
RNA template by stopping and reinitiating at each gene junction and transcribes genes in a
sequential and gradient manner. Accordingly the first gene, NP, is transcribed at the highest
level whereas the last gene, L is transcribed at the lowest level. Then, replication likely
begins when enough NP is present to encapsidate neo-synthetized antigenome and
genomes. GP-encoding mRNAs transit to the endoplasmic reticulum (ER) where GP is
synthesized and form trimers. After the addition of N and O-linked glycans in the ER and
Golgi apparatus, GPs are delivered to the plasma membrane by secretory vesicles. NP, VP35
and VP30 proteins associate with viral RNA to form RNP complex, and with matrix proteins
(VP40 and VP24) and GP proteins. Eventually, viral particles bud at the cell surface and are
released.
Figure 5 : EBOV life cycle, from White JM 2012 A new player in the puzzle of filovirus entry [49]
10
III - Pathogenesis
1 - Target cells and tissues
The detailed pathogenesis of the disease is not well understood. Nevertheless, it has been
found that EBOV has a broad cell tropism, infecting a wide range of cell types. In situ
hybridization and electron microscopy analyses of tissues from patients with fatal disease or
from experimentally infected non-human primates showed that monocytes, macrophages,
dendritic cells (DCs), endothelial cells, fibroblasts and several types of epithelial cells such as
hepatocytes and adrenal cortical cells support EBOV replication [50-54]. Temporal in vivo
studies in non-human primates experimentally infected with EBOV determined that
monocytes, macrophages, DCs but also natural killer (NK) cells are the first and favorite
targets of the virus, whereas all others cells cited above are infected much later during the
course of the disease, proximal to death [51, 52, 55]. Monocytes, macrophages, and DCs
appear to play a major role in the dissemination of the virus. Immunohistochemical studies
have shown that the virus disseminates from lymph nodes via lymphatic and vascular
systems to several organs including liver, spleen, lung, kidney, pancreas, large and small
intestines and skin amongst others [50, 54]. Nevertheless, the most prominent damages are
observed in liver and spleen. In these organs, cell necrosis and apoptosis were detected. The
same was observed in lymph nodes leading to the lymphoid depletion detailed below see
chapter IV.2). In the liver, hepatocytes and Kupffer cells are infected, leading to hepatic
dysfunction directly resulting from viral damages or circulatory impairment. EBOV infection
leads to coagulopathy through damages to both liver, which is the production site of clotting
factors, as well as certain coagulation inhibitors, [56] and endothelial cells, which provide
tissue factor (TF also known as thromboplastin), tissue factor pathway inhibitor (TFPI) and
receptor for protein C activation [50, 57]. These organ disorders contribute more to the
patient death than the hemorrhagic fever.
2 - Coagulation anomalies and vascular endothelium impact
Coagulopathy has been observed during EBOV infection and might have several causes
including activation of cytokine secretion, platelet aggregation and consumption, activation
of the coagulation cascade, deficiency of coagulation factors due to both liver and
endothelium damages. Indeed, it has been described that pro-inflammatory cytokines such
as IL-6 are increased in human and non-human primates infected by EBOV [58, 59]. IL-6 is
known to trigger the coagulation cascade. Accordingly, the transcriptional targets of IL-6
including several proteins that either increase the transcription of pro-coagulant proteins
like TF or decrease the transcription of anticoagulant proteins such as antithrombin [60].
Moreover, EBOV infected monocytes and macrophages induce an increase of TF protein
level in macaques circulation [61]. EBOV infection also causes hepatic necrosis and apoptosis
leading to an impairment of the synthesis of critical coagulation factor including protein C,
11
protein S and fibrinogen [62, 63]. Deregulation of this coagulation pathway leads to
disseminated intravascular coagulation (DIC) which is observed during infection and likely
contributes to hemorrhage symptoms and multi-organ failure [6, 61, 64]. In addition to these
problems in the coagulation pathway, the widespread injury to endothelial cells via a direct
cytotoxic effect of GP (detailed later in chapter III.3) is observed in EBOV infection and is
another mechanism triggering DIC. These cells have several properties, one of these being
the capacity to regulate the process of coagulation and fibrinolysis and to modulate the
fibrin deposition. At steady state, endothelial cell surface is thought to be essentially
anticoagulant or non-thrombogenic. The control of coagulation is exerted by endothelial
cells at different critical steps of the clotting cascade. Briefly, endothelial cells are the main
source of TFPI, which blocks TF, the principal initiator of the coagulation cascade [65, 66]. TF
is a transmembrane glycoprotein receptor expressed in response to injury at the surface of a
variety of cells, including platelets, monocytes, macrophages, fibroblasts, and endothelial
cells [67]. In addition, endothelial cells express a large amount of heparan sulfate and related
glycosaminoglycans to neutralized clotting enzymes such as factor Xa and thrombin [65].
Eventually, endothelial cells play a critical role in the protein C anticoagulant pathway by
deregulating its expression [68]. EBOV infection leads to impairment of the endothelial
barrier integrity and to an increased endothelial permeability [51]. In addition, several
factors secreted by both infected monocytes and macrophages can exert changes in the
vascular endothelium in a variety of ways. This includes either an indirect induction of
endothelial cell activation, by infecting and activating leukocytes and triggering the synthesis
and local production of pro-inflammatory soluble factors, or a direct induction of changes in
endothelial cell expression of cytokines, chemokines and cell adhesion molecules in the
absence of immune mediators (as a direct result of virus infection, mechanism detailed in
chapter III.3). Mediators released from EBOV-activated endothelial cells can modulate
vascular tone, thrombosis, and/or inflammation including nitric oxide (NO), prostacyclin,
interferons (IFNs), interleukin (IL)-1, IL-6, and chemokines such as IL-8, IL-6, IL-7 [61]. All
these endothelial cells impairments are implicated in DIC syndrome and hemorrhage
development. However, as previously mentioned, the hemorrhage observed during EBOV
infection is insufficient to cause the death, as the massive loss of blood is atypical and, when
is present, is largely restricted to the gastrointestinal tract. Nevertheless, it seems that pro-
inflammatory cytokines secreted by monocytes, macrophages or DCs and both apoptosis
and necrosis observed in several organs including liver and spleen induced by EBOV infection
might participate to malfunction of both vascular system and coagulation, leading to general
failure of several organs, septic shock and death.
12
3 - Direct toxicity
In vitro studies have shown that GP has direct cytotoxic properties on endothelial cells via
morphology changes leading to cell rounding and detachment [69, 70]. Indeed, studies
identified a reduction at the cell surface of the expression of adhesion molecules such as
integrins or immune molecules (including major histocompatibility complex class I [MHC]
and the epidermal growth factor receptor) induced by GP expression. This is believed to
contribute to the cell rounding and consequent loss of cell adhesion observed in infected
cells [70-74]. This finding suggests that GP and more particularly the MLR of GP plays an
important role in endothelial cell toxicity and could be responsible for both endothelial
integrity disruption and increased endothelial permeability, triggering hemorrhage
development during the disease [69]. The mechanism by which GP has this toxic effect has
been shown to be dependent on GTPase dynamin. Through its interaction with dynamin, GP
disrupts the normal intracellular trafficking of the cell surface proteins essential for cell
attachment and immune signaling [70]. Nevertheless, the importance of GP cytotoxicity in
viral pathogenesis is however controversial. Indeed, direct damages to the endothelial cells
by virus replication have been observed only in animal models at terminal stages of the
disease [51]. A study demonstrated that moderate expression of GP (similar to the amount
observed in EBOV infected cells during the early stages of infection) did not result in
morphological changes and was not cytotoxic, suggesting that cell rounding and
downregulation of the surface markers are late events in EBOV infection, whereas
production and massive release of virus particles occur at early steps [75].
It has been described that EBOV-infected cells release proteolytic endosomal enzymes, such
as the cathepsin proteases implicated in extracellular matrix degradation and disease
progression [76, 77]. The secretion of cathepsins by EBOV-infected cells suggests that these
molecules may be implicated in direct cytotoxicity induced by EBOV and contribute to the
vascular endothelium destruction because these proteases in vitro catalyze the degradation
of extracellular matrix and induce cell rounding and detachment in vitro.
A recent study showed that GP increases the NK cell toxicity. In fact, mouse macrophages
infected with VSV particles containing EBOV-GP instead of their glycoprotein (VSVΔG/EBOV-
GP) particles causes an increase in NK cell cytotoxicity through a decrease of MHC-I
expression [55].
13
IV - Immune response during EBOV infection
Several studies have shown that EBOV infection was associated with aberrant innate
immune responses and with global suppression of adaptive immunity (Figure 6).
1 - Innate response
After the epithelial barrier, innate immunity is defined as the first line of defense against
pathogenic microbial exposure. Innate immune responses are not specific to a particular
pathogen in contrast to the adaptive immune responses. Innate immune responses involve
several pathways in order to distinguish self from non-self. The recognition of non-self leads
to the activation of several cells, such as monocytes, macrophages, granulocytes, DCs,
natural killer (NK) cells but also to the complement activation (soluble factors) followed by
the cytokines production such as interferon (IFN). The interferon system represents a major
innate defense against infections by viruses and other pathogens. Three classes of IFNs have
been described. Type I IFNs, comprising IFN-α and IFN-β, are produced by many cell types.
Type II IFNs, with IFN-γ, are generated by activated T cells and NK cells. Type III IFNs,
including IFN λ1–3, are incompletely characterized, but are believed to mediate an antiviral
response as well. The IFN response begins with the recognition of diverse pathogen-
associated molecular patterns (PAMPs) by pattern recognition receptors (PRRs). Viruses
contain several PAMPs recognized by specific PRRs. Double strand RNA (dsRNA), single
strand RNA, CpG-DNA, 5'-triphosphate RNA or single strand DNA which are recognized by
Toll-like receptors (TLR)-3 , TLR 7/8, TLR9, retinoic acid-inducible gene I (RIG-I) or single
stranded DNA melanoma differentiation associated gene 5 (MDA-5). EBOV, being a negative
strand RNA, induces IFN signaling through TLR-3, TLR-7/8 and RIG-I. The receptors can be
localized in the cytoplasm, like RIG-I and MDA-5, or in membranes, like TLRs. Receptor
activation leads to IFN production via IFN regulatory factor (IRF)-3 and IRF-7. This secreted
IFN binds to its receptor composed of two subunits, IFN α receptor 1 (IFNAR1) and IFNAR2 at
the cell surface in order to activate JAK-STAT pathway. This signaling pathway leads to the
phosphorylation and subsequent dimerization of the transcription factor STAT, allowing its
shuttle into the nucleus to induce transcription of interferon-stimulated genes (ISG)[78]. IFN
response leads to an antiviral state but we will see later that several EBOV proteins can
interfere at several levels with the JAK/STAT pathway (see chapter V.2). Altogether, these
mechanisms are often sufficient to counter invading viruses. In addition, when they fail to do
so, they favor the generation of host mediated humoral and cellular immune responses that
limit and in most cases eliminate the invading pathogen. Several viruses, however, such as
EBOV, have developed a variety of mechanisms to escape the innate immune system
(detailed below). EBOV infection targets antigen-presenting cells (APC) during the early
stages of infection. Since these cells play a critical role in immune responses, their infection
by EBOV has dramatic consequences, notably by preventing their maturation. Indeed, in
14
vitro studies demonstrated that EBOV-infected DCs do not express the DC
maturation/activation markers such as CD80, CD86, CD40, CD83 and MHC of class I and II
needed to CD4+
and CD8+
T-cell co-simulation and activation [79, 80]. In addition, EBOV
infection prevents cytokine and chemokine production implicated in inflammation
regulation and immune response such as IFN-α, IFN-β, tumor necrosis factor (TNF) -α, IL-1β,
IL-10, IL-6, IL-2, IL-8, IL-12 [79, 81]. During viral infection, NK cells quickly respond by
triggering exocytosis of perforin and granzymes and secretion of IFN-γ, respectively
mediating the destruction of infected cells or the macrophage activation. NK cells activation
requires several signaling molecules including IL-12 (for the cytokines production), IFN-α and
IFN-β (for the development of cytotoxic effector function) secreted by mature/activated
DCs. Since EBOV infection prevents DC maturation/activation, NK cells activation is
decreased [82] , which further favors virus replication. Therefore, a proper activation of NK
cells could be critical for the protection of EBOV infection [83].
In such a dysregulated immune response context, it has been observed that despite a high
viral load and necrotic lesions in fatal EBOV cases, only a minimal inflammation is observed
in infected organs and tissues [50], probably due to a weak immune system activation. Yet,
and in contrast to the negative impact of EBOV infection on DCs and NK cells, the infection of
monocytes and macrophages by EBOV leads to an important secretion of pro-inflammatory
cytokines such as IL-1β, IL-6, IL-8, IL-15, IL-16, TNF-α but not IFN-α and chemokines such as
macrophage inflammatory protein (MIP)- 1α, MIP-β [84-86]. All these disturbances in
immune cell activation and pro- and anti-inflammatory cytokines production contribute to
facilitate the uncontrolled viral replication observed during EBOV infection. Indeed, it has
been shown that the early innate response correlates with the survival of EBOV-infected
patients. Therefore, the rapid initiation of innate response may limit EBOV infection and
could be a critical condition to host survival [32].
2 - Adaptive response
Adaptive response is the third line of defense, after epithelial barrier and innate response. It
is triggered after a few days of infection and is more powerful than innate immunity in
combating the infection. In contrast with innate system, adaptive system develops a specific
response to the antigen and allows establishing an immune memory. Briefly, after DC
maturation and activation by pathogen detection, DCs migrate to lymph nodes to present
antigens on their surface via MHC-I or II and express co-stimulatory factors (CD80, CD84,
CD40) in order to activate T lymphocytes (CD4+ and CD8+). When pathogen-specific T cells
are activated, they proliferate, leave the lymph node and migrate to infected tissues. CD8+ T
cells directly kill the infected cells through their cytotoxic activity and CD4+ T cells (Th1)
activate macrophages via both TCR-MHC-II interaction and cytokine production. Another
CD4+ T cells (Th2) population remains in the lymph node and stimulates the proliferation
and differentiation of pathogen-specific B cells through both MHC-II presentation and
15
cytokine production in order to promote the antigen-specific antibody production or
proliferation of memory B cells.
We have seen that EBOV replicates efficiently in DCs without cytokine and chemokine
production and without inducing their maturation/activation. This lack of DC activation most
likely results into poor immune responses by NK as seen before but also into weak T and B
cell activation. In addition, fatal cases of EBOV infection are associated with a lack of
detectable adaptive immunity. It has been observed that EBOV infection induces a
substantial lymphopenia due to CD4+ and CD8+ T cell depletion and necrosis observed at
least in spleen, thymus and lymph nodes of non survivors compared to survivors; the same
was observed in experimentally infected non-human primates [50, 87, 88], and the different
mechanisms implicated in this phenomenon will be described below (paragraph
"lymphopenia"). Nevertheless, despite significant lymphocyte apoptosis, it has been
demonstrated that a functional and specific, albeit insufficient, adaptive immune response is
present in lethal EBOV infection [89], occurring even in the presence of incompletely
activated DCs. There is an increased percentage of CD4+ and CD8+ T cells expressing high
levels of CD44, a T-cell activation and maturation marker, close to the end of lethal EBOV
infection. CD8+ T cells play an important role in EBOV infection. Indeed, in lethal mice model
of EBOV, the IFN-γ production by CD8+ T cells in response to EBOV infection was observed at
the end of the disease [89]. In addition, this important source of IFN-γ could explain the
macrophage and monocyte activation observed during EBOV infection. Moreover, transfer
of EBOV-specific CD8+ T cells from mice infected with EBOV during 7 days protects naive
mice from EBOV challenge. [89]. Together, these data support the hypothesis that functional
adaptive immune responses are present, at the end of the disease in lethal EBOV-infected
mice but is insufficient in part due to massive lymphocyte apoptosis.
Concerning B cells, a clinical study performed during the 1996 outbreak in Gabon described
humoral immune responses in EBOV infected patients, as antibodies directed against GP
have been found in surviving patients [90]. In addition, important levels of IgG and IgM,
specific to NP, VP40 and VP35, have been found by ELISA in all survivors early in disease or
during early convalescence. In contrast, no viral antigen-specific IgG have been found in fatal
cases and only weak IgM levels have been detected in one-third of fatal cases [33]. These
results suggest that a prompt and vigorous humoral response may help survivors to limit and
finally control viral dissemination. Furthermore, it has been observed that this
immunoglobulin deficiency is not associated with a decrease of B cells. The mechanism by
which EBOV impacts on immunoglobulin levels therefore remains poorly understood [33,
91]. Nevertheless, the impact of EBOV infection on T cell activation and proliferation could
alter B cell activation.
16
Lymphopenia
The mechanism by which EBOV induces a lymphopenia is not fully understood, likely in part
because a direct mechanism cannot be involved since EBOV does not infect lymphocytes.
As discussed above, EBOV readily infects and replicates in DCs, interfering with their
activation/maturation and therefore with their ability to initiate the adaptive immune
response and the associated lymphocytes expansion [61, 80].
We have seen before that the release of NO, which is a physiological vasodilator and anti-
platelet factor, was increased by the endothelial cells activated by EBOV [61, 92]. In vivo
study demonstrated that blood levels of NO were much higher in fatal cases (increasing with
disease severity), and extremely elevated levels could have negatively affected vascular tone
and contributed to virus-induced shock [93]. In addition of its role in endothelial barrier, NO
could also play a role in lymphopenia. Briefly, it has been shown that NO promotes apoptotic
pathways in numerous cell types including lymphocytes through the indirect activation of
caspases [94], moreover NO inhibits T and B cell proliferation via the downregulation of
MHC-II, co-stimulation molecules and/or cytokines (such as IL-12) [95].
The death receptor pathway activation could be implicated in lymphocyte apoptosis
observed during EBOV infection. EBOV infection could induce both intrinsic (mitochondrial
mediated pathway) and extrinsic (death receptor pathway) cell death cascades as crosstalk
occurs between these two pathways. In the intrinsic pathway, intracellular stress factors
(such as oxidative stress or DNA injury) via Bax and Bak proteins cause depolarization of the
mitochondrial membrane, thereby inducing the release of cytochrome C and activating the
caspase cascade beginning with caspase-9. Bcl-2 protein is known to inhibit apoptosis via its
interaction with Bax and Bak proteins [96]. In fatal EBOV cases, a decrease of Bcl-2 mRNA
level has been observed in PBMC during the disease, whereas a strong increase has been
detected in survivors at the time of T-cell activation [33]. The extrinsic pathway is initiated
by ligand-receptor interaction at the cell surface including either Fas Ligand with Fas or TRAIL
with TRAIL receptors (such as DR4 and DR5). Such interactions lead to caspase (caspase 8
and then caspases 3 and 7) activation via the adaptor protein Fas-associated death domain
(FADD) and finally induce DNA degradation and cell death [97]. With regards to the extrinsic
pathway, EBOV infection increases TRAIL expression in cultured monocyte-like cells, and
some EBOV-infected monkeys exhibit an increase of soluble Fas in their sera [86].
Furthermore, TRAIL and Fas mRNA expressions are increased in the PBMC of infected
monkeys [52].
Another explanation for lymphopenia induced by EBOV implicates GP. Indeed GP contains a
domain with a significant homology with the "immunosuppressive peptide" found in
glycoproteins of various oncogenic retroviruses known to often induce immunosuppression
[98]. This will be detailed below (see chapter V.1.c). In addition, it was postulated that sGP
17
could play a role in lymphocyte apoptosis by interacting with circulating lymphocytes, as it
was detected in large amounts in the blood [99]. Nevertheless, an in vivo study showed that
sGP was not able to induce T cell apoptosis neither by itself nor by death receptor co-
stimulation; further studies are required to investigate the ability of sGP to induce apoptosis
via the intrinsic pathway [100].
Altogether, immune system dysfunctions (weak innate and adaptive immune system
activation or lymphopenia) contribute to the uncontrolled spread and growth of the virus.
This suggests that a strong immune response may result in protection against EBOV
infection, which may guide the design of new therapeutic strategies to control lethal EBOV
disease.
Figure 6 : Model of EBOV pathogenesis in primates. Adapted to Bray M 2005 [101]
18
V - Immune response evasion by EBOV viral proteins
Several mechanisms by which EBOV escapes immune system have been suggested (Figure
7).
Figure 7 : Potential mechanisms by which various EBOV proteins evade host innate and acquired immune systems.
Adapted from Ansari AA 2014 [102]
1 - GP implications
a - Glycosylation and MLR
EBOV infection elicits only low level of neutralizing antibodies against GP in humans and
other animals [14]. As mentioned above, the heavy glycosylation of GP is implicated in the
immune system escape. These glycans located in the MLR sequence promote the generation
of antibodies against the more variable GP1 domain, which are not able to confer a strong
protection [103]. In mice, removal of the MLR of GP1 can lead to the production of more
efficient antibodies directed against the conserved glycoprotein core structure, confirming
the impact of this MLR domain in masking neutralizing epitopes [104]. Moreover, O- and N-
glycosylations impedes the recognition of GP by neutralizing antibodies through steric
shielding [41, 74, 105-107]. In addition, MLR is necessary and sufficient to decrease the
expression of cell surface proteins such as MHC-I and several members of the integrin family.
This domain blocks the access to MHC-I needed for CD8+ T cell stimulation [69, 105]. An
additional mechanism by which glycosylations play an important function in the immune
escape involves N-linked GP2 glycosylation. Indeed, mutation of one of the two N-linked GP2
glycosylation sites prevents the interaction between GP1 and GP2 required for GP
localization at the plasma membrane and is implicated in antigenicity and immunogenicity of
EBOV GP. All these results suggest that it might be possible to enhance immunity by specific
modifications in the GP glycosylation [37].
19
In addition, we have seen that EBOV infection leads to DC maturation defects and
consequently to a failure of efficient T cell activation [79, 80]. In vitro studies using EBOV-
virus like particles (VLP) containing VP40 demonstrated that VLPs, contrary to EBOV
infection, have the capacity to activate DCs. MLR is the domain required for DC activation via
a recognition of MLR by toll like receptor (TLR)-4 and NF-kappaB and MAPK signaling
pathway activation [108, 109]. Indeed, VLPs with wild-type GP but not with MLR-deleted GP
can activate TLR-4-dependent responses. In EBOV infection context, these results suggest
that MLR plays a major role in the abnormal DC activation observed during the disease.
b - Role of sGP and shed GP
Secreted GPs, sGP and shed GP, have been shown to be important in immune evasion.
Because sGP shares 295 amino acids with GP and is the predominant transcript for the GP
gene, it has been postulated that sGP probably competes with virion-attached GP. Indeed,
the majority of antibodies from EBOV-surviving patients and monkeys are directed against
sGP rather than against GP1/2 [110, 111]. It is possible that the majority of antibodies
binding sGP are non-neutralizing, but it is likely that the weak amount of neutralizing
antibody production is absorbed by the much more abundant sGP. Indeed, it has been
demonstrated that sGP serves as a decoy for neutralizing antibodies [112]. In addition to its
role in adaptive response evasion, sGP has been reported to bind to neutrophils through the
Fcγ receptor thereby inhibiting early neutrophil activation [113]. Concerning the shed GP, in
a guinea pig model of EBOV infection, this secreted GP is present in significant amounts in
the blood of infected animals. Shed GP inhibits the neutralizing activity of EBOV antibodies,
and the increase of shed GP in infected animals observed between days 6 and 9 post
infection correlates with the course of disease and the lethal outcome at day 9 [114]. All
these findings suggest that secreted GPs may play an important role in the pathogenesis.
c - Immunosuppressive domain in GP
Several retroviruses including Avian reticuloendotheliosis virus (ARV) and Feline leukemia
virus (FeLV), have a particular peptide in their envelope protein named p15E or
"immunosuppressive peptide", that has immunosuppressive properties. For example, this
peptide inhibits the T cell activation normally induced by concanavalin stimulation [115], the
proliferation of murine cytotoxic T cells [116] and macrophage recruitment to the
inflammatory site in mice [117]. Amino acid sequence comparison has uncovered a high
homology between this "immunosuppressive peptide" and 160 residues at the C-terminal
part of EBOV-GP which could explain the immunosuppressive effect mediated by EBOV-GP
[98]. More recently, a study identified a 17-mer peptide in this region as the
immunosuppressive domain of EBOV-GP. This peptide induces a significant decline of CD4+
and CD8+ T cells. In addition, this peptide induces a decrease of IL-2 receptor at the T cell
surface, but also inhibits IFN-γ, IL-2 and IL-10 expression leading to an inhibition of T cell
proliferation and activation [118]. The mechanism by which immunosuppressive peptide
20
acts on CD4+ and CD8+ cells is unknown but it has been hypothesized that it inactivates
these cells by directly contacting them or indirectly through its previously described effect
on APCs.
2 - IFN pathway inhibition by VP35 and VP24
EBOV uses several mechanisms in order to inhibit IFN production (Figure 8). It has been
shown that VP35 is responsible for the absence of IFN-α production and prevents the
activation of IFN-stimulated response element (ISRE)-containing promoters when either
transfected dsRNA or viral infection is used as the activating stimulus [119]. A more detailed
study of the mechanism by which VP35 influences the host IFN response showed that it
inhibits the IFN synthesis at several levels. VP35 can bind viral dsRNA and inhibit the
recognition by helicase RIG-I implicated in the IFN pathway and then the IFN-α and -β
production [120]. The ability of VP35 to block IFN production was also correlated with its
ability to inhibit the phosphorylation of IRF-3 through interaction with kinases including IκB
kinase epsilon (IKKε) and TANK-binding kinase 1 (TBK-1) [121], and thus inhibiting its nuclear
translocation and activation [122]. A SUMOylation of IRF-7 induced by VP35 was recently
described as an additional mechanism of repression of the transcription of IFN genes [123].
Co-immunoprecipitation experiments demonstrated that VP35 interacts with PIAS1 (protein
inhibitor of activated STAT-1) and Ubc9, two proteins involved in the small ubiquitin-like
modifier (SUMO) conjugation cascade [124, 125]. Besides that, Feng et al have
demonstrated that VP35 protein is a RNA binding protein with a stronger affinity for dsRNA
than PKR. Consequently, VP35 competes with PKR for EBOV dsRNA binding and prevents the
phosphorylation of translation initiation factor eIF-2 (eIF-2) by PKR required to stop protein
synthesis and thus viral replication [126].
In addition to VP35, VP24 is another important player in the counteraction of IFN pathway
by EBOV. VP24 inhibits the IFN pathway by preventing the nuclear accumulation of STAT-1
[127]. Actually, VP24 binds to karyopherin-α, a nuclear transporter, with very high affinity to
compete with STAT-1 and inhibit its nuclear transport [128, 129]. In addition to the JAK-STAT
pathway, the p38 mitogen-activated protein (MAP) kinase pathway is also critical for the IFN
response [130]. Engagement of the IFN receptor by IFN activates a cascade of MAP kinases,
leading to the phosphorylation of the alpha isoform of p38 (p38-α) [131]. Phosphorylated
p38-α then triggers the phosphorylation of downstream transcription factors that participate
in IFN responses. It is well established that p38 is essential for gene transcription via ISRE or
GAS elements [130-132]. It has been observed that VP24 inhibits the p38 MAP kinase
pathway by preventing the phosphorylation of p38-α [133]. The dual action of these two
viral proteins, VP35 and VP24, may thus contribute to a potent inhibition of the IFN pathway,
permitting an efficient virus replication and dissemination in the host.
21
Figure 8 : EBOV proteins interfering with interferon signaling
22
VI - Diagnosis and treatments
Although EBOV is considered to be a significant public health problem, no licensed drug or
vaccine is currently available [134-136]. The most effective measure for controlling disease
propagation is the isolation of patients and strict barrier nursing procedures to protect
healthcare workers. Meanwhile, symptomatic and supportive care is the treatment of
choice. Nevertheless, owing to the advances of basic EBOV research, several promising drugs
and vaccine candidates [137] are under development.
1 - Diagnosis methods
As written above, the clinical symptoms in the early stages of EBOV infection are very similar
to others viral diseases such as flu and other respiratory infections, common enteritis or
other infections frequently occurring in African including malaria and Lassa fever. Therefore,
especially in the early stages, virological testing is very important for the diagnosis.
Specific EBOV-antibodies detection by ELISA and immunofluorescence has been developed
but as mentioned above, EBOV antibodies are produced only in small quantity, especially in
fatal cases.
The inoculation of a cell culture with patient sera or other body fluids or tissue extracts is the
classical method to isolate and amplify EBOV. Then, EBOV is detected by PCR or
immunofluorescence using viral-specific primers or antibodies respectively. Antigen blood
tests are based on the detection of virus proteins using specific antibodies and are hardly
influenced by virus variability. The high viremia in EBOV patients often facilitates antigen
detection, although the tests are clinically less sensitive than PCR [138, 139]. As EBOV has a
specific filamentous morphology, the direct detection of EBOV by electron microscopy in
organ section and serum is possible but high virus concentrations are needed [140]. A major
disadvantage of these diagnosis methods is the time required to isolate the virus (days to
week) and the need of biosafety level 3 or 4 facilities. The detection by electronic
microscopy is not routinely used because of its high cost. Therefore, the most used method
is based on nucleic acid tests, as it requires 24-48h to obtain results in a very sensitive
fashion. Very recently a new test that provides results within 15 minutes has been
developed, the ReEBOVTM
Antigen Rapid Test. This test, which is based on the detection of
the VP40 protein rather than nucleic acids, is able to correctly identify about 92% of EBOV
infected patients and to exclude 85% of those not infected with the virus. In addition to its
rapidity, the antigen test is easy to perform and does not require electricity, which therefore
would favor its use in lower health care facilities or mobile units [141].
23
2 - Treatments and Vaccines
Currently, the majority of treatments used aim at treating symptoms induced by EBOV. For
example, as EBOV inhibits IFN signaling, exogenous INF-α or INF-β have been used and could
delay the occurrence of viremia or increase survival time, but they cannot rescue non-
human primates from lethal infection [142, 143]. As EBOV infection indirectly impairs the
coagulation pathway by provoking the depletion of clotting factors through aberrant and
excessive coagulation, the recombinant nematode anticoagulant protein c2 (rNAPc2) and
the recombinant human activated protein C (rhAPC), originally used for anticoagulation
purposes, have been tested and gave promising results in infected monkey [63, 144].
rNAPc2, which has shown 33% efficacy in non-human primates [144], is in Phase II trial for
thrombosis prevention. Nevertheless no human trial is planned for EBOV treatment [145].
Several treatments targeting a specific step of viral life cycle including entry, RNA synthesis
and translation have been developed.
a - Candidates to block the viral entry
In order to block the virus entry, researchers purified patients-derived polyclonal or
monoclonal antibodies specifically targeting the main neutralizing epitopes on EBOV-GP. The
antibody KZ52, derived from a survivor of the Kikwit EBOV outbreak in 1995, displays a
potent neutralizing activity and has been shown to protect guinea pigs [146] but not non-
human primates [147]. During the past years, researchers have developed three generations
of antibody cocktail formulations. The first one was based on the combination of two
human-mouse chimeric antibodies, ch133 and ch226, which presented strong neutralizing
activity against EBOV in vitro. Unfortunately, trials in non-human primates challenged with
EBOV were not convincing [148]. A second generation of anti-EBOV antibody cocktail
formulas, ZMAb and MB-003 consisting of three different neutralizing antibodies derived
from EBOV GP, have been tested in non-human primates. ZMAb, containing mAbs 1H3, 2G4
and 4G7, showed 100% protection in Cynomolgus macaques [149]. The MB-003 cocktail,
including antibodies of c13C6, h-13F6, and c6D8, showed 67% protection in macaques [150].
It seems that human trial has started so far for this treatment. This technology may be
insufficiently robust to promote the production of neutralizing antibodies to fight the
current EBOV outbreak. A recent study has established a better optimized antibody
combination derived Zmab and MB-003, named Zmapp and containing c13C6, 2G4 and 4G7.
This new mAbs combination demonstrates a successful protection in non-human primates
[151]. Phase I safety and efficacy trials have been initiated in January 2015, but the
conclusions are not yet available [145].
In addition to the neutralizing antibodies, other treatments have been developed to block
viral entry. Since the first C-terminal heptad repeat (CHR)-peptide-based HIV entry inhibitor
24
discovered in 1992 [152], this potential treatment strategy has been applied against many
enveloped viruses, including EBOV [153, 154]. Briefly, as the CHR domain of GP2 plays a role
during the fusion step in the endosomes, exogenous CHR could be able to compete with viral
CHR and prevent the viral fusion. This treatment showed inhibition activity against three
EBOV species, including Zaire, Sudan and Reston Ebolavirus [153]. Other therapeutic
candidates have been described to prevent the fusion step including Cat L/B inhibitor [155]
and NPC binding compounds [156].
b - Candidates to block viral RNA synthesis and/or translation
Others drugs targeting RNA synthesis and translation have been developed. Nucleot(s)ides
analogues including Ribavirin, Favipiravir and Brincidofovir have been tested. Ribavirin could
not limit the replication of EBOV and failed to protect animals from lethal challenge [157,
158]. Interestingly, Favipiravir showed efficient antiviral activity in mouse models for EBOV
infections [159]. Clinical efficacy trials began in Guinea in December 2014, however more
data are required in order to draw a conclusion [145]. Brincidofovir (CMX001), showed
potent anti-EBOV activity in vitro, and has been used to treat EBOV patients but its
mechanism of action is unclear. However, a new phase II clinical trials of Brincidofovir has
been launched to test its potential safety and antiviral activity in EBOV infected patients
[160]. A new clinical efficacy trial began in Liberia in January 2015, but due to the lack of
patients this trial has been stopped. In addition, to date no precise results are available
because this drug is often combined with other drug therapies [145]. Finally, BCX-4430,
another nucleoside analogue, interferes with the function of RNA polymerase of EBOV, and
confers protection to EBOV-challenged rodent animals [161]. BCX-4430 is in phase I safety
trial and efficacy trials will begin providing that the safety results from Phase I will be
satisfactory [145].
Others strategies using small interfering RNAs (siRNAs) have been developed. Especially,
siRNAs specifically directed against the RNA sequences of RNP complex, VP24, and VP35
were tested [162]. For instance AVI-6002, a mixture of iRNA targeting mRNA sequences of
VP24 and VP35 protected five of eight rhesus monkeys from EBOV challenge [163]. For this
drug, the phase I safety is completed but there are no human trial planned at this time [145].
c - Vaccines
Several vaccine candidates have been tested on rodent and non-human primates [164]. The
first trials were done with inactivated viruses but this method was quickly abandoned. A lot
of viral vectors have been used to produced anti-EBOV vaccines including Venezuelan equine
encephalitis virus [165], adenovirus [166], virus Parainfluenza [167] or Vesicular stomatitis
virus (VSV) [168].
Attenuated recombinant VSV vaccine expressing EBOV GP protects non-human primates
from EBOV infection. Interestingly, it has been used to successfully treat a scientist infected
25
by EBOV [169]. Clinical trials are in progress in several countries including United States,
Canada, Germany, Gabon and Switzerland. Concerning the last one, clinical trials are
performed in Geneva and began in September 2014. Initial data obtained were very
promising but the development of unexpected mild to moderate joint pain 10 to 15 days
after injection had lead to the suspension of this trial. In January 2015, the trial resumed
using a lower dose and final results are expected soon.
The appearance of reverse genetic tools for EBOV allowed the opening of a new way in the
design of vaccine vectors. For example, it has been shown that EBOV recombinant carrying
mutations in the domain of the VP35 involved in the suppression of IFN production loses its
virulence in a guinea pig model [170]. It also effectively protects guinea pigs during EBOV
infection. However, this method is unsafe since the recombinant EBOV could mutate and
therby regain its pathogenic potential in the vaccinated patient. VLPs expressing
immunogenic proteins such as the NP, GP and VP40 EBOV were also tested [171], but this
approach is expensive and difficult to implement.
VII - Conclusion
This review summarizes the major knowledge on EBOV accumulated during almost 40 years.
Unfortunately, the entry receptors, virus life cycle, immune response and evasion during
infection are not fully understood. As of today, there are no vaccines or efficient treatment
available. However, this virus has caused a lot of deaths since 1976. But the interest for the
research even if it seems to correlate with death cases (Figure S2), was scanty for many
years probably due to the fact that outbreaks spread only in Africa, and thus far away from
Western countries. Interestingly, two cases of Marburg virus (a virus close to EBOV and with
similar symptoms) have been detected in 2008 in the Netherlands [172] and the United
States [173]. These cases alerted the international community on the risk of emergent viral
diseases and have had a positive impact on the number of publications related to EBOV
(Figure S2). In addition, the ongoing outbreak, has caused a huge increase of publications on
EBOV (Figure S3). After almost 40 years and thousands of deaths, EBOV finally begins to
receive some attention from researchers, and more precisely from the organizations that
fund basic research and the pharmacological companies.
26
References
1. Kuhn, J.H., et al., Virus nomenclature below the species level: a standardized nomenclature
for natural variants of viruses assigned to the family Filoviridae. Arch Virol, 2013. 158(1): p.
301-11.
2. Kuhn, J.H., et al., Proposal for a revised taxonomy of the family Filoviridae: classification,
names of taxa and viruses, and virus abbreviations. Arch Virol, 2010. 155(12): p. 2083-103.
3. Carroll, S.A., et al., Molecular evolution of viruses of the family Filoviridae based on 97 whole-
genome sequences. J Virol, 2013. 87(5): p. 2608-16.
4. Baize, S., et al., Emergence of Zaire Ebola virus disease in Guinea. N Engl J Med, 2014.
371(15): p. 1418-25.
5. Ebola haemorrhagic fever in Sudan, 1976. Report of a WHO/International Study Team. Bull
World Health Organ, 1978. 56(2): p. 247-70.
6. Ebola haemorrhagic fever in Zaire, 1976. Bull World Health Organ, 1978. 56(2): p. 271-93.
7. WHO. Ebola virus disease, Disease outbreak news. 2014.
8. OMS. Maladie à virus Ebola. 2014.
9. WHO. Ebola Situation Report - 8 March 2015. 2015.
10. Leroy, E.M., et al., Fruit bats as reservoirs of Ebola virus. Nature, 2005. 438(7068): p. 575-6.
11. Nkoghe, D., et al., [Practical guidelines for the management of Ebola infected patients in the
field]. Med Trop (Mars), 2004. 64(2): p. 199-204.
12. Neumann, G., S. Watanabe, and Y. Kawaoka, Characterization of Ebolavirus regulatory
genomic regions. Virus Res, 2009. 144(1-2): p. 1-7.
13. Lee, J.E. and E.O. Saphire, Ebolavirus glycoprotein structure and mechanism of entry. Future
Virol, 2009. 4(6): p. 621-635.
14. Feldmann, H.S., A. Geisbert, TW., Filoviridae: Marburg and Ebola Viruses, in Fileds Virology,
D.H. Knipe, PM., Editor. 2013. p. 923-956.
15. Jeffers, S.A., D.A. Sanders, and A. Sanchez, Covalent modifications of the ebola virus
glycoprotein. J Virol, 2002. 76(24): p. 12463-72.
16. Mackay, I.M. Ebola virus disease: obliterating a variant and stalled case decline... 2015.
17. Huang, Y., et al., The assembly of Ebola virus nucleocapsid requires virion-associated proteins
35 and 24 and posttranslational modification of nucleoprotein. Mol Cell, 2002. 10(2): p. 307-
16.
18. Martinez, M.J., et al., Role of Ebola virus VP30 in transcription reinitiation. J Virol, 2008.
82(24): p. 12569-73.
19. Scianimanico, S., et al., Membrane association induces a conformational change in the Ebola
virus matrix protein. EMBO J, 2000. 19(24): p. 6732-41.
20. Hoenen, T., et al., VP40 octamers are essential for Ebola virus replication. J Virol, 2005. 79(3):
p. 1898-905.
21. Dessen, A., et al., Crystal structure of the matrix protein VP40 from Ebola virus. EMBO J, 2000.
19(16): p. 4228-36.
22. Ruigrok, R.W., et al., Structural characterization and membrane binding properties of the
matrix protein VP40 of Ebola virus. J Mol Biol, 2000. 300(1): p. 103-12.
23. Bornholdt, Z.A., et al., Structural rearrangement of ebola virus VP40 begets multiple functions
in the virus life cycle. Cell, 2013. 154(4): p. 763-74.
24. Adu-Gyamfi, E., et al., Investigation of Ebola VP40 assembly and oligomerization in live cells
using number and brightness analysis. Biophys J, 2012. 102(11): p. 2517-25.
25. Gomis-Ruth, F.X., et al., The matrix protein VP40 from Ebola virus octamerizes into pore-like
structures with specific RNA binding properties. Structure, 2003. 11(4): p. 423-33.
26. Timmins, J., et al., Oligomerization and polymerization of the filovirus matrix protein VP40.
Virology, 2003. 312(2): p. 359-68.
27
27. Adu-Gyamfi, E., et al., The Ebola virus matrix protein penetrates into the plasma membrane:
a key step in viral protein 40 (VP40) oligomerization and viral egress. J Biol Chem, 2013.
288(8): p. 5779-89.
28. Jasenosky, L.D., et al., Ebola virus VP40-induced particle formation and association with the
lipid bilayer. J Virol, 2001. 75(11): p. 5205-14.
29. Noda, T., et al., Ebola virus VP40 drives the formation of virus-like filamentous particles along
with GP. J Virol, 2002. 76(10): p. 4855-65.
30. Soni, S.P., et al., The Ebola virus matrix protein deeply penetrates the plasma membrane: an
important step in viral egress. Biophys J, 2013. 104(9): p. 1940-9.
31. Reynard, O., et al., Unconventional secretion of Ebola virus matrix protein VP40. J Infect Dis,
2011. 204 Suppl 3: p. S833-9.
32. Leroy, E.M., et al., Early immune responses accompanying human asymptomatic Ebola
infections. Clin Exp Immunol, 2001. 124(3): p. 453-60.
33. Baize, S., et al., Defective humoral responses and extensive intravascular apoptosis are
associated with fatal outcome in Ebola virus-infected patients. Nat Med, 1999. 5(4): p. 423-6.
34. Watt, A., et al., A novel life cycle modeling system for Ebola virus shows a genome length-
dependent role of VP24 in virus infectivity. J Virol, 2014. 88(18): p. 10511-24.
35. Volchkov, V.E., et al., Processing of the Ebola virus glycoprotein by the proprotein convertase
furin. Proc Natl Acad Sci U S A, 1998. 95(10): p. 5762-7.
36. Lee, J.E., et al., Structure of the Ebola virus glycoprotein bound to an antibody from a human
survivor. Nature, 2008. 454(7201): p. 177-82.
37. Dowling, W., et al., Influences of glycosylation on antigenicity, immunogenicity, and
protective efficacy of ebola virus GP DNA vaccines. J Virol, 2007. 81(4): p. 1821-37.
38. Volchkov, V.E., et al., Recovery of infectious Ebola virus from complementary DNA: RNA
editing of the GP gene and viral cytotoxicity. Science, 2001. 291(5510): p. 1965-9.
39. Sanchez, A., et al., Detection and molecular characterization of Ebola viruses causing disease
in human and nonhuman primates. J Infect Dis, 1999. 179 Suppl 1: p. S164-9.
40. Iwasa, A., M. Shimojima, and Y. Kawaoka, sGP serves as a structural protein in Ebola virus
infection. J Infect Dis, 2011. 204 Suppl 3: p. S897-903.
41. Cook, J.D. and J.E. Lee, The secret life of viral entry glycoproteins: moonlighting in immune
evasion. PLoS Pathog, 2013. 9(5): p. e1003258.
42. Matsuno, K., et al., C-type lectins do not act as functional receptors for filovirus entry into
cells. Biochem Biophys Res Commun, 2010. 403(1): p. 144-8.
43. Kondratowicz, A.S., et al., T-cell immunoglobulin and mucin domain 1 (TIM-1) is a receptor for
Zaire Ebolavirus and Lake Victoria Marburgvirus. Proc Natl Acad Sci U S A, 2011. 108(20): p.
8426-31.
44. Moller-Tank, S., et al., Role of the phosphatidylserine receptor TIM-1 in enveloped-virus entry.
J Virol, 2013. 87(15): p. 8327-41.
45. Aleksandrowicz, P., et al., Ebola virus enters host cells by macropinocytosis and clathrin-
mediated endocytosis. J Infect Dis, 2011. 204 Suppl 3: p. S957-67.
46. Gnirss, K., et al., Cathepsins B and L activate Ebola but not Marburg virus glycoproteins for
efficient entry into cell lines and macrophages independent of TMPRSS2 expression. Virology,
2012. 424(1): p. 3-10.
47. Ng, M., et al., Cell entry by a novel European filovirus requires host endosomal cysteine
proteases and Niemann-Pick C1. Virology, 2014. 468-470: p. 637-46.
48. Carette, J.E., et al., Ebola virus entry requires the cholesterol transporter Niemann-Pick C1.
Nature, 2011. 477(7364): p. 340-3.
49. White, J.M. and K.L. Schornberg, A new player in the puzzle of filovirus entry. Nat Rev
Microbiol, 2012. 10(5): p. 317-22.
28
50. Martines, R.B., et al., Tissue and cellular tropism, pathology and pathogenesis of Ebola and
Marburg viruses. J Pathol, 2015. 235(2): p. 153-74.
51. Geisbert, T.W., et al., Pathogenesis of Ebola hemorrhagic fever in primate models: evidence
that hemorrhage is not a direct effect of virus-induced cytolysis of endothelial cells. Am J
Pathol, 2003. 163(6): p. 2371-82.
52. Geisbert, T.W., et al., Pathogenesis of Ebola hemorrhagic fever in cynomolgus macaques:
evidence that dendritic cells are early and sustained targets of infection. Am J Pathol, 2003.
163(6): p. 2347-70.
53. Zaki, S.R., 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: p. S36-47.
54. Ryabchikova, E.I., L.V. Kolesnikova, and S.V. Luchko, An analysis of features of pathogenesis
in two animal models of Ebola virus infection. J Infect Dis, 1999. 179 Suppl 1: p. S199-202.
55. Williams, K.J., et al., VSVDeltaG/EBOV GP-Induced Innate Protection Enhances Natural Killer
Cell Activity to Increase Survival in a Lethal Mouse Adapted Ebola Virus Infection. Viral
Immunol, 2015. 28(1): p. 51-61.
56. Blonski, W., T. Siropaides, and K.R. Reddy, Coagulopathy in liver disease. Curr Treat Options
Gastroenterol, 2007. 10(6): p. 464-73.
57. van Hinsbergh, V.W., Endothelium--role in regulation of coagulation and inflammation. Semin
Immunopathol, 2012. 34(1): p. 93-106.
58. Hensley, L.E. and T.W. Geisbert, The contribution of the endothelium to the development of
coagulation disorders that characterize Ebola hemorrhagic fever in primates. Thromb
Haemost, 2005. 94(2): p. 254-61.
59. Leroy, E.M., et al., Human asymptomatic Ebola infection and strong inflammatory response.
Lancet, 2000. 355(9222): p. 2210-5.
60. Kerr, R., D. Stirling, and C.A. Ludlam, Interleukin 6 and haemostasis. Br J Haematol, 2001.
115(1): p. 3-12.
61. Geisbert, T.W., et al., Mechanisms underlying coagulation abnormalities in ebola
hemorrhagic fever: overexpression of tissue factor in primate monocytes/macrophages is a
key event. J Infect Dis, 2003. 188(11): p. 1618-29.
62. Ebihara, H., et al., Host response dynamics following lethal infection of rhesus macaques with
Zaire ebolavirus. J Infect Dis, 2011. 204 Suppl 3: p. S991-9.
63. Hensley, L.E., et al., Recombinant human activated protein C for the postexposure treatment
of Ebola hemorrhagic fever. J Infect Dis, 2007. 196 Suppl 2: p. S390-9.
64. Zampieri, C.A., N.J. Sullivan, and G.J. Nabel, Immunopathology of highly virulent pathogens:
insights from Ebola virus. Nat Immunol, 2007. 8(11): p. 1159-64.
65. Bombeli, T., M. Mueller, and A. Haeberli, Anticoagulant properties of the vascular
endothelium. Thromb Haemost, 1997. 77(3): p. 408-23.
66. Broze, G.J., Jr., Tissue factor pathway inhibitor. Thromb Haemost, 1995. 74(1): p. 90-3.
67. Chu, A.J., Tissue factor, blood coagulation, and beyond: an overview. Int J Inflam, 2011. 2011:
p. 367284.
68. Esmon, C.T., The anticoagulant and anti-inflammatory roles of the protein C anticoagulant
pathway. J Autoimmun, 2000. 15(2): p. 113-6.
69. Yang, Z.Y., et al., Identification of the Ebola virus glycoprotein as the main viral determinant
of vascular cell cytotoxicity and injury. Nat Med, 2000. 6(8): p. 886-9.
70. Sullivan, N.J., et al., Ebola virus glycoprotein toxicity is mediated by a dynamin-dependent
protein-trafficking pathway. J Virol, 2005. 79(1): p. 547-53.
71. Simmons, G., et al., Ebola virus glycoproteins induce global surface protein down-modulation
and loss of cell adherence. J Virol, 2002. 76(5): p. 2518-28.
29
72. Escudero-Perez, B., et al., Shed GP of Ebola virus triggers immune activation and increased
vascular permeability. PLoS Pathog, 2014. 10(11): p. e1004509.
73. Ray, R.B., et al., Ebola virus glycoprotein-mediated anoikis of primary human cardiac
microvascular endothelial cells. Virology, 2004. 321(2): p. 181-8.
74. Reynard, O., et al., Ebolavirus glycoprotein GP masks both its own epitopes and the presence
of cellular surface proteins. J Virol, 2009. 83(18): p. 9596-601.
75. Alazard-Dany, N., et al., Ebola virus glycoprotein GP is not cytotoxic when expressed
constitutively at a moderate level. J Gen Virol, 2006. 87(Pt 5): p. 1247-57.
76. Barrientos, L.G. and P.E. Rollin, Release of cellular proteases into the acidic extracellular
milieu exacerbates Ebola virus-induced cell damage. Virology, 2007. 358(1): p. 1-9.
77. Nomura, T. and N. Katunuma, Involvement of cathepsins in the invasion, metastasis and
proliferation of cancer cells. J Med Invest, 2005. 52(1-2): p. 1-9.
78. Schoggins, J.W., et al., A diverse range of gene products are effectors of the type I interferon
antiviral response. Nature, 2011. 472(7344): p. 481-5.
79. Bosio, C.M., et al., Ebola and Marburg viruses replicate in monocyte-derived dendritic cells
without inducing the production of cytokines and full maturation. J Infect Dis, 2003. 188(11):
p. 1630-8.
80. Mahanty, S., et al., Cutting edge: impairment of dendritic cells and adaptive immunity by
Ebola and Lassa viruses. J Immunol, 2003. 170(6): p. 2797-801.
81. Yen, B., et al., Molecular basis for ebolavirus VP35 suppression of human dendritic cell
maturation. J Virol, 2014. 88(21): p. 12500-10.
82. Alli, R.S. and A. Khar, Interleukin-12 secreted by mature dendritic cells mediates activation of
NK cell function. FEBS Lett, 2004. 559(1-3): p. 71-6.
83. Warfield, K.L., et al., Role of natural killer cells in innate protection against lethal ebola virus
infection. J Exp Med, 2004. 200(2): p. 169-79.
84. Gupta, M., et al., Monocyte-derived human macrophages and peripheral blood mononuclear
cells infected with ebola virus secrete MIP-1alpha and TNF-alpha and inhibit poly-IC-induced
IFN-alpha in vitro. Virology, 2001. 284(1): p. 20-5.
85. Stroher, U., et al., Infection and activation of monocytes by Marburg and Ebola viruses. J
Virol, 2001. 75(22): p. 11025-33.
86. Hensley, L.E., et al., Proinflammatory response during Ebola virus infection of primate models:
possible involvement of the tumor necrosis factor receptor superfamily. Immunol Lett, 2002.
80(3): p. 169-79.
87. Wauquier, N., et al., Human fatal zaire ebola virus infection is associated with an aberrant
innate immunity and with massive lymphocyte apoptosis. PLoS Negl Trop Dis, 2010. 4(10).
88. Bradfute, S.B., et al., Lymphocyte death in a mouse model of Ebola virus infection. J Infect Dis,
2007. 196 Suppl 2: p. S296-304.
89. Bradfute, S.B., K.L. Warfield, and S. Bavari, Functional CD8+ T cell responses in lethal Ebola
virus infection. J Immunol, 2008. 180(6): p. 4058-66.
90. Maruyama, T., et al., Ebola virus can be effectively neutralized by antibody produced in
natural human infection. J Virol, 1999. 73(7): p. 6024-30.
91. Reed, D.S., et al., Depletion of peripheral blood T lymphocytes and NK cells during the course
of ebola hemorrhagic Fever in cynomolgus macaques. Viral Immunol, 2004. 17(3): p. 390-400.
92. Yang, Y. and J. Loscalzo, Regulation of tissue factor expression in human microvascular
endothelial cells by nitric oxide. Circulation, 2000. 101(18): p. 2144-8.
93. Sanchez, A., et al., Analysis of human peripheral blood samples from fatal and nonfatal cases
of Ebola (Sudan) hemorrhagic fever: cellular responses, virus load, and nitric oxide levels. J
Virol, 2004. 78(19): p. 10370-7.
94. Kim, P.K., et al., Regulation of caspases by nitric oxide. Ann N Y Acad Sci, 2002. 962: p. 42-52.
95. Bogdan, C., Nitric oxide and the immune response. Nat Immunol, 2001. 2(10): p. 907-16.
30
96. Kroemer, G., The proto-oncogene Bcl-2 and its role in regulating apoptosis. Nat Med, 1997.
3(6): p. 614-20.
97. Lavrik, I., A. Golks, and P.H. Krammer, Death receptor signaling. J Cell Sci, 2005. 118(Pt 2): p.
265-7.
98. Volchkov, V.E., V.M. Blinov, and S.V. Netesov, The envelope glycoprotein of Ebola virus
contains an immunosuppressive-like domain similar to oncogenic retroviruses. FEBS Lett,
1992. 305(3): p. 181-4.
99. Sanchez, A., et al., The virion glycoproteins of Ebola viruses are encoded in two reading
frames and are expressed through transcriptional editing. Proc Natl Acad Sci U S A, 1996.
93(8): p. 3602-7.
100. Wolf, K., et al., The Ebola virus soluble glycoprotein (sGP) does not affect lymphocyte
apoptosis and adhesion to activated endothelium. J Infect Dis, 2011. 204 Suppl 3: p. S947-52.
101. Bray, M. and T.W. Geisbert, Ebola virus: the role of macrophages and dendritic cells in the
pathogenesis of Ebola hemorrhagic fever. Int J Biochem Cell Biol, 2005. 37(8): p. 1560-6.
102. Ansari, A.A., Clinical features and pathobiology of Ebolavirus infection. J Autoimmun, 2014.
55: p. 1-9.
103. Wilson, J.A., et al., Epitopes involved in antibody-mediated protection from Ebola virus.
Science, 2000. 287(5458): p. 1664-6.
104. Ou, W., et al., Induction of ebolavirus cross-species immunity using retrovirus-like particles
bearing the Ebola virus glycoprotein lacking the mucin-like domain. Virol J, 2012. 9: p. 32.
105. Francica, J.R., et al., Steric shielding of surface epitopes and impaired immune recognition
induced by the ebola virus glycoprotein. PLoS Pathog, 2010. 6(9): p. e1001098.
106. Marzi, A., et al., Antibodies are necessary for rVSV/ZEBOV-GP-mediated protection against
lethal Ebola virus challenge in nonhuman primates. Proc Natl Acad Sci U S A, 2013. 110(5): p.
1893-8.
107. Wong, G., et al., Immune parameters correlate with protection against ebola virus infection in
rodents and nonhuman primates. Sci Transl Med, 2012. 4(158): p. 158ra146.
108. Okumura, A., et al., Interaction between Ebola virus glycoprotein and host toll-like receptor 4
leads to induction of proinflammatory cytokines and SOCS1. J Virol, 2010. 84(1): p. 27-33.
109. Martinez, O., C. Valmas, and C.F. Basler, Ebola virus-like particle-induced activation of NF-
kappaB and Erk signaling in human dendritic cells requires the glycoprotein mucin domain.
Virology, 2007. 364(2): p. 342-54.
110. Druar, C., et al., Analysis of the expressed heavy chain variable-region genes of Macaca
fascicularis and isolation of monoclonal antibodies specific for the Ebola virus' soluble
glycoprotein. Immunogenetics, 2005. 57(10): p. 730-8.
111. Maruyama, T., et al., Recombinant human monoclonal antibodies to Ebola virus. J Infect Dis,
1999. 179 Suppl 1: p. S235-9.
112. Ito, H., et al., Ebola virus glycoprotein: proteolytic processing, acylation, cell tropism, and
detection of neutralizing antibodies. J Virol, 2001. 75(3): p. 1576-80.
113. Yang, Z., et al., Distinct cellular interactions of secreted and transmembrane Ebola virus
glycoproteins. Science, 1998. 279(5353): p. 1034-7.
114. Dolnik, O., et al., Ectodomain shedding of the glycoprotein GP of Ebola virus. EMBO J, 2004.
23(10): p. 2175-84.
115. Copelan, E.A., et al., The mechanism of retrovirus suppression of human T cell proliferation in
vitro. J Immunol, 1983. 131(4): p. 2017-20.
116. Orosz, C.G., et al., Retrovirus-mediated immunosuppression. I. FeLV-UV and specific FeLV
proteins alter T lymphocyte behavior by inducing hyporesponsiveness to lymphokines. J
Immunol, 1985. 134(5): p. 3396-403.
117. Cianciolo, G.J., et al., Macrophage accumulation in mice is inhibited by low molecular weight
products from murine leukemia viruses. J Immunol, 1980. 124(6): p. 2900-5.
31
118. Yaddanapudi, K., et al., Implication of a retrovirus-like glycoprotein peptide in the
immunopathogenesis of Ebola and Marburg viruses. FASEB J, 2006. 20(14): p. 2519-30.
119. Basler, C.F., et al., The Ebola virus VP35 protein functions as a type I IFN antagonist. Proc Natl
Acad Sci U S A, 2000. 97(22): p. 12289-94.
120. Cardenas, W.B., et al., Ebola virus VP35 protein binds double-stranded RNA and inhibits
alpha/beta interferon production induced by RIG-I signaling. J Virol, 2006. 80(11): p. 5168-78.
121. Prins, K.C., W.B. Cardenas, and C.F. Basler, Ebola virus protein VP35 impairs the function of
interferon regulatory factor-activating kinases IKKepsilon and TBK-1. J Virol, 2009. 83(7): p.
3069-77.
122. Basler, C.F., et al., The Ebola virus VP35 protein inhibits activation of interferon regulatory
factor 3. J Virol, 2003. 77(14): p. 7945-56.
123. Chang, T.H., et al., Ebola Zaire virus blocks type I interferon production by exploiting the host
SUMO modification machinery. PLoS Pathog, 2009. 5(6): p. e1000493.
124. Hay, R.T., SUMO: a history of modification. Mol Cell, 2005. 18(1): p. 1-12.
125. Meulmeester, E. and F. Melchior, Cell biology: SUMO. Nature, 2008. 452(7188): p. 709-11.
126. Feng, Z., et al., The VP35 protein of Ebola virus inhibits the antiviral effect mediated by
double-stranded RNA-dependent protein kinase PKR. J Virol, 2007. 81(1): p. 182-92.
127. Reid, S.P., et al., Ebola virus VP24 binds karyopherin alpha1 and blocks STAT1 nuclear
accumulation. J Virol, 2006. 80(11): p. 5156-67.
128. Xu, W., et al., Ebola virus VP24 targets a unique NLS binding site on karyopherin alpha 5 to
selectively compete with nuclear import of phosphorylated STAT1. Cell Host Microbe, 2014.
16(2): p. 187-200.
129. Reid, S.P., et al., Ebola virus VP24 proteins inhibit the interaction of NPI-1 subfamily
karyopherin alpha proteins with activated STAT1. J Virol, 2007. 81(24): p. 13469-77.
130. Platanias, L.C., Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev
Immunol, 2005. 5(5): p. 375-86.
131. Uddin, S., et al., Activation of the p38 mitogen-activated protein kinase by type I interferons. J
Biol Chem, 1999. 274(42): p. 30127-31.
132. Platanias, L.C., The p38 mitogen-activated protein kinase pathway and its role in interferon
signaling. Pharmacol Ther, 2003. 98(2): p. 129-42.
133. Halfmann, P., G. Neumann, and Y. Kawaoka, The Ebolavirus VP24 protein blocks
phosphorylation of p38 mitogen-activated protein kinase. J Infect Dis, 2011. 204 Suppl 3: p.
S953-6.
134. Hoenen, T., A. Groseth, and H. Feldmann, Current ebola vaccines. Expert Opin Biol Ther,
2012. 12(7): p. 859-72.
135. Marzi, A. and H. Feldmann, Ebola virus vaccines: an overview of current approaches. Expert
Rev Vaccines, 2014. 13(4): p. 521-31.
136. Mullard, A., Experimental Ebola drugs enter the limelight. Lancet, 2014. 384(9944): p. 649.
137. Ye, L. and C. Yang, Development of vaccines for prevention of Ebola virus infection. Microbes
Infect, 2015. 17(2): p. 98-108.
138. Bausch, D.G., et al., Diagnosis and clinical virology of Lassa fever as evaluated by enzyme-
linked immunosorbent assay, indirect fluorescent-antibody test, and virus isolation. J Clin
Microbiol, 2000. 38(7): p. 2670-7.
139. Leroy, E.M., et al., Diagnosis of Ebola haemorrhagic fever by RT-PCR in an epidemic setting. J
Med Virol, 2000. 60(4): p. 463-7.
140. Biel, S.S. and H.R. Gelderblom, Diagnostic electron microscopy is still a timely and rewarding
method. J Clin Virol, 1999. 13(1-2): p. 105-19.
141. WHO, WHO Emergency Use Assessment and Listing for Ebola Virus Disease IVDs, Product:
ReEBOV™ Antigen Rapid Test Kit. Febuary 2015, WHO.
32
142. Jahrling, P.B., et al., Evaluation of immune globulin and recombinant interferon-alpha2b for
treatment of experimental Ebola virus infections. J Infect Dis, 1999. 179 Suppl 1: p. S224-34.
143. Smith, L.M., et al., Interferon-beta therapy prolongs survival in rhesus macaque models of
Ebola and Marburg hemorrhagic fever. J Infect Dis, 2013. 208(2): p. 310-8.
144. Geisbert, T.W., et al., Treatment of Ebola virus infection with a recombinant inhibitor of
factor VIIa/tissue factor: a study in rhesus monkeys. Lancet, 2003. 362(9400): p. 1953-8.
145. WHO, Categorization and prioritization of drugs for consideration for testing or use in
patients infected with Ebola. 2015, World Health Organization:
http://who.int/medicines/ebola-treatment/cat_prioritization_drugs_testing/en/.
146. Parren, P.W., et al., Pre- and postexposure prophylaxis of Ebola virus infection in an animal
model by passive transfer of a neutralizing human antibody. J Virol, 2002. 76(12): p. 6408-12.
147. Oswald, W.B., et al., Neutralizing antibody fails to impact the course of Ebola virus infection
in monkeys. PLoS Pathog, 2007. 3(1): p. e9.
148. Marzi, A., et al., Protective efficacy of neutralizing monoclonal antibodies in a nonhuman
primate model of Ebola hemorrhagic fever. PLoS One, 2012. 7(4): p. e36192.
149. Qiu, X., et al., Successful treatment of ebola virus-infected cynomolgus macaques with
monoclonal antibodies. Sci Transl Med, 2012. 4(138): p. 138ra81.
150. Olinger, G.G., Jr., et al., Delayed treatment of Ebola virus infection with plant-derived
monoclonal antibodies provides protection in rhesus macaques. Proc Natl Acad Sci U S A,
2012. 109(44): p. 18030-5.
151. Qiu, X., et al., Reversion of advanced Ebola virus disease in nonhuman primates with ZMapp.
Nature, 2014. 514(7520): p. 47-53.
152. Jiang, S., et al., HIV-1 inhibition by a peptide. Nature, 1993. 365(6442): p. 113.
153. Miller, E.H., et al., Inhibition of Ebola virus entry by a C-peptide targeted to endosomes. J Biol
Chem, 2011. 286(18): p. 15854-61.
154. Higgins, C.D., et al., C-peptide inhibitors of Ebola virus glycoprotein-mediated cell entry:
effects of conjugation to cholesterol and side chain-side chain crosslinking. Bioorg Med Chem
Lett, 2013. 23(19): p. 5356-60.
155. Shah, P.P., et al., A small-molecule oxocarbazate inhibitor of human cathepsin L blocks severe
acute respiratory syndrome and ebola pseudotype virus infection into human embryonic
kidney 293T cells. Mol Pharmacol, 2010. 78(2): p. 319-24.
156. Cote, M., et al., Small molecule inhibitors reveal Niemann-Pick C1 is essential for Ebola virus
infection. Nature, 2011. 477(7364): p. 344-8.
157. Canonico, P.G., et al., In-vivo activity of antivirals against exotic RNA viral infections. J
Antimicrob Chemother, 1984. 14 Suppl A: p. 27-41.
158. Huggins, J.W., Prospects for treatment of viral hemorrhagic fevers with ribavirin, a broad-
spectrum antiviral drug. Rev Infect Dis, 1989. 11 Suppl 4: p. S750-61.
159. Smither, S.J., et al., Post-exposure efficacy of oral T-705 (Favipiravir) against inhalational
Ebola virus infection in a mouse model. Antiviral Res, 2014. 104: p. 153-5.
160. Chimerix, I. Brincidofovir for Ebola. Chimerix, Inc. 2014 [cited 22.11.14.
161. Warren, T.K., et al., Protection against filovirus diseases by a novel broad-spectrum
nucleoside analogue BCX4430. Nature, 2014. 508(7496): p. 402-5.
162. Geisbert, T.W., et al., Postexposure protection of guinea pigs against a lethal ebola virus
challenge is conferred by RNA interference. J Infect Dis, 2006. 193(12): p. 1650-7.
163. Warren, T.K., et al., Advanced antisense therapies for postexposure protection against lethal
filovirus infections. Nat Med, 2010. 16(9): p. 991-4.
164. Grant-Klein, R.J., L.A. Altamura, and C.S. Schmaljohn, Progress in recombinant DNA-derived
vaccines for Lassa virus and filoviruses. Virus Res, 2011. 162(1-2): p. 148-61.
165. Pushko, P., et al., Individual and bivalent vaccines based on alphavirus replicons protect
guinea pigs against infection with Lassa and Ebola viruses. J Virol, 2001. 75(23): p. 11677-85.
33
166. Sullivan, N.J., et al., Accelerated vaccination for Ebola virus haemorrhagic fever in non-human
primates. Nature, 2003. 424(6949): p. 681-4.
167. Bukreyev, A., et al., Successful topical respiratory tract immunization of primates against
Ebola virus. J Virol, 2007. 81(12): p. 6379-88.
168. Garbutt, M., et al., Properties of replication-competent vesicular stomatitis virus vectors
expressing glycoproteins of filoviruses and arenaviruses. J Virol, 2004. 78(10): p. 5458-65.
169. Feldmann, H., et al., Effective post-exposure treatment of Ebola infection. PLoS Pathog, 2007.
3(1): p. e2.
170. Prins, K.C., et al., Mutations abrogating VP35 interaction with double-stranded RNA render
Ebola virus avirulent in guinea pigs. J Virol, 2010. 84(6): p. 3004-15.
171. Warfield, K.L., et al., Ebola virus-like particle-based vaccine protects nonhuman primates
against lethal Ebola virus challenge. J Infect Dis, 2007. 196 Suppl 2: p. S430-7.
172. WHO. Case of Marburg Haemorrhagic Fever imported into the Netherlands from Uganda.
2008.
173. CDC, C.f.D.C.a.P. Imported Case of Marburg Hemorrhagic Fever --- Colorado, 2008 2009.
174. Science-Metrix. L’épidémie du virus Ebola et les publications scientifiques – La communauté
scientifique réagit rapidement! 2014.
34
Supplementary information:
Figure S1 : Worldwide geographic distribution of filovirus hemorrhagic fever cases, 1967–2014. From Martines et all
(2015) [50]
35
Figure S2 : Ebola fatal cases and scientific publications on Ebola, 1976- 2014 [174]
Figure S3 : Ebola fatal cases and scientific publications on Ebola, March 2014 - October 2014 [174]

More Related Content

What's hot

SEMINAR PPT PRESENTATION
SEMINAR PPT PRESENTATIONSEMINAR PPT PRESENTATION
SEMINAR PPT PRESENTATION
JERRY ABU
 
Oncogenic viruses
Oncogenic virusesOncogenic viruses
Oncogenic viruses
Balamurugan r
 
Oncogenic dna virus
Oncogenic dna virus Oncogenic dna virus
John Short dissertation poster
John Short dissertation posterJohn Short dissertation poster
John Short dissertation poster
John Alexander Logan Short
 
human oncogenic viruses
human oncogenic viruseshuman oncogenic viruses
human oncogenic viruses
SHUBHANGI2509
 
40. tv
40. tv40. tv
oncogenic viruses ppt.
oncogenic viruses ppt.oncogenic viruses ppt.
oncogenic viruses ppt.
DrRavi Bhushan
 
Oncogenic virus-host cell interaction
Oncogenic virus-host cell interactionOncogenic virus-host cell interaction
Oncogenic virus-host cell interaction
Mousumi Bora
 
Rift Valley fever in East Africa: Factors driving emergence, potential interv...
Rift Valley fever in East Africa: Factors driving emergence, potential interv...Rift Valley fever in East Africa: Factors driving emergence, potential interv...
Rift Valley fever in East Africa: Factors driving emergence, potential interv...
ILRI
 
Human Endogenous Retrovirus (power point)
Human Endogenous Retrovirus (power point)Human Endogenous Retrovirus (power point)
Human Endogenous Retrovirus (power point)
Kevin B Hugins
 
Infections and cancer
Infections and cancerInfections and cancer
Infections and cancer
Dr. Amer Ali Khaleel /HMU
 
Oncogenic virus ppt
Oncogenic virus pptOncogenic virus ppt
Oncogenic virus ppt
Deepak Sarangi
 
Human endogenous retroviruses
Human endogenous retroviruses Human endogenous retroviruses
Human endogenous retroviruses
naeim1370
 
Rota
RotaRota
Potyviridae
PotyviridaePotyviridae
Potyviridae
UdayenduL10
 
AIDS
AIDSAIDS
Oncogenic Viruses
Oncogenic VirusesOncogenic Viruses
Oncogenic Viruses
Dr. Rakesh Prasad Sah
 
Gutell 063.jmb.1997.269.0203
Gutell 063.jmb.1997.269.0203Gutell 063.jmb.1997.269.0203
Gutell 063.jmb.1997.269.0203
Robin Gutell
 
Human t lymphotropic viruses
Human t lymphotropic virusesHuman t lymphotropic viruses
Human t lymphotropic viruses
Rena Faith Baradero
 
Infectious diseases and control strategies in shrimp
Infectious diseases and control strategies in shrimpInfectious diseases and control strategies in shrimp
Infectious diseases and control strategies in shrimp
International Aquafeed
 

What's hot (20)

SEMINAR PPT PRESENTATION
SEMINAR PPT PRESENTATIONSEMINAR PPT PRESENTATION
SEMINAR PPT PRESENTATION
 
Oncogenic viruses
Oncogenic virusesOncogenic viruses
Oncogenic viruses
 
Oncogenic dna virus
Oncogenic dna virus Oncogenic dna virus
Oncogenic dna virus
 
John Short dissertation poster
John Short dissertation posterJohn Short dissertation poster
John Short dissertation poster
 
human oncogenic viruses
human oncogenic viruseshuman oncogenic viruses
human oncogenic viruses
 
40. tv
40. tv40. tv
40. tv
 
oncogenic viruses ppt.
oncogenic viruses ppt.oncogenic viruses ppt.
oncogenic viruses ppt.
 
Oncogenic virus-host cell interaction
Oncogenic virus-host cell interactionOncogenic virus-host cell interaction
Oncogenic virus-host cell interaction
 
Rift Valley fever in East Africa: Factors driving emergence, potential interv...
Rift Valley fever in East Africa: Factors driving emergence, potential interv...Rift Valley fever in East Africa: Factors driving emergence, potential interv...
Rift Valley fever in East Africa: Factors driving emergence, potential interv...
 
Human Endogenous Retrovirus (power point)
Human Endogenous Retrovirus (power point)Human Endogenous Retrovirus (power point)
Human Endogenous Retrovirus (power point)
 
Infections and cancer
Infections and cancerInfections and cancer
Infections and cancer
 
Oncogenic virus ppt
Oncogenic virus pptOncogenic virus ppt
Oncogenic virus ppt
 
Human endogenous retroviruses
Human endogenous retroviruses Human endogenous retroviruses
Human endogenous retroviruses
 
Rota
RotaRota
Rota
 
Potyviridae
PotyviridaePotyviridae
Potyviridae
 
AIDS
AIDSAIDS
AIDS
 
Oncogenic Viruses
Oncogenic VirusesOncogenic Viruses
Oncogenic Viruses
 
Gutell 063.jmb.1997.269.0203
Gutell 063.jmb.1997.269.0203Gutell 063.jmb.1997.269.0203
Gutell 063.jmb.1997.269.0203
 
Human t lymphotropic viruses
Human t lymphotropic virusesHuman t lymphotropic viruses
Human t lymphotropic viruses
 
Infectious diseases and control strategies in shrimp
Infectious diseases and control strategies in shrimpInfectious diseases and control strategies in shrimp
Infectious diseases and control strategies in shrimp
 

Viewers also liked

Visual Rhetoric Project
Visual Rhetoric ProjectVisual Rhetoric Project
Visual Rhetoric Project
Anna del Villar
 
Fessell Improv Article
Fessell Improv ArticleFessell Improv Article
Fessell Improv Article
David Fessell
 
Strengths Analysis
Strengths AnalysisStrengths Analysis
Strengths Analysis
Nick Graff
 
Centro de acondicionamiento J.C.
Centro de acondicionamiento J.C.Centro de acondicionamiento J.C.
Centro de acondicionamiento J.C.
maria camila diaz mesa
 
Positioning strategies of two wheeler companie
Positioning strategies of two wheeler companie Positioning strategies of two wheeler companie
Positioning strategies of two wheeler companie
DEEPAK VERMA
 
Sitharthan_CV
Sitharthan_CVSitharthan_CV
Sitharthan_CV
SITHARTHAN MANI
 
6a chearstenwebbpowerpoint
6a chearstenwebbpowerpoint6a chearstenwebbpowerpoint
6a chearstenwebbpowerpoint
Chearsten Webb
 
abstract of thesis
abstract of thesisabstract of thesis
abstract of thesis
Emilie Branche
 
shyam_infor SunSystem Analyst
shyam_infor SunSystem Analyst  shyam_infor SunSystem Analyst
shyam_infor SunSystem Analyst
Shyam Pokharkar
 
Looma_HMI(executive)_content WxH
Looma_HMI(executive)_content WxHLooma_HMI(executive)_content WxH
Looma_HMI(executive)_content WxH
Wei (Vicky) Guo
 
NJ 2015 Year in Review
NJ 2015 Year in ReviewNJ 2015 Year in Review
NJ 2015 Year in Review
Andrew Perrotti
 
Clases de cuerpos geometricos
Clases de cuerpos geometricosClases de cuerpos geometricos
Clases de cuerpos geometricos
INMA ARCE SANCHEZ
 
Colloqium
ColloqiumColloqium

Viewers also liked (13)

Visual Rhetoric Project
Visual Rhetoric ProjectVisual Rhetoric Project
Visual Rhetoric Project
 
Fessell Improv Article
Fessell Improv ArticleFessell Improv Article
Fessell Improv Article
 
Strengths Analysis
Strengths AnalysisStrengths Analysis
Strengths Analysis
 
Centro de acondicionamiento J.C.
Centro de acondicionamiento J.C.Centro de acondicionamiento J.C.
Centro de acondicionamiento J.C.
 
Positioning strategies of two wheeler companie
Positioning strategies of two wheeler companie Positioning strategies of two wheeler companie
Positioning strategies of two wheeler companie
 
Sitharthan_CV
Sitharthan_CVSitharthan_CV
Sitharthan_CV
 
6a chearstenwebbpowerpoint
6a chearstenwebbpowerpoint6a chearstenwebbpowerpoint
6a chearstenwebbpowerpoint
 
abstract of thesis
abstract of thesisabstract of thesis
abstract of thesis
 
shyam_infor SunSystem Analyst
shyam_infor SunSystem Analyst  shyam_infor SunSystem Analyst
shyam_infor SunSystem Analyst
 
Looma_HMI(executive)_content WxH
Looma_HMI(executive)_content WxHLooma_HMI(executive)_content WxH
Looma_HMI(executive)_content WxH
 
NJ 2015 Year in Review
NJ 2015 Year in ReviewNJ 2015 Year in Review
NJ 2015 Year in Review
 
Clases de cuerpos geometricos
Clases de cuerpos geometricosClases de cuerpos geometricos
Clases de cuerpos geometricos
 
Colloqium
ColloqiumColloqium
Colloqium
 

Similar to Branche-Hors these

JSResEssay15-Final-TCDkparsad.doc
JSResEssay15-Final-TCDkparsad.docJSResEssay15-Final-TCDkparsad.doc
JSResEssay15-Final-TCDkparsad.doc
Kanchan Parsad
 
EBOLA & OTHER VHF 2022.pptx
EBOLA & OTHER VHF 2022.pptxEBOLA & OTHER VHF 2022.pptx
EBOLA & OTHER VHF 2022.pptx
Mkindi Mkindi
 
Ebola.pptx
Ebola.pptxEbola.pptx
Ebola.pptx
BinteHawah1
 
Ebola virus disease - A comprehensive review
Ebola virus disease - A comprehensive reviewEbola virus disease - A comprehensive review
Ebola virus disease - A comprehensive review
pharmaindexing
 
Ebola Viruse slid show.docx
Ebola Viruse slid show.docxEbola Viruse slid show.docx
Ebola Viruse slid show.docx
AlizaAshraf3
 
Genomic sm musa marena (1)
Genomic sm musa marena (1)Genomic sm musa marena (1)
Genomic sm musa marena (1)
marenam
 
Ebola virus a to z
Ebola virus a to zEbola virus a to z
Ebola virus a to z
Ashok Babu Naidu Yakkateela
 
EBOLA VIRUS
EBOLA VIRUSEBOLA VIRUS
EBOLA VIRUS
Sachin Shekde
 
Analysis Of Avian Avulavirus
Analysis Of Avian AvulavirusAnalysis Of Avian Avulavirus
Analysis Of Avian Avulavirus
Camella Taylor
 
Ebola virus complete ppt students of jcpt -m.b
Ebola virus complete ppt students of jcpt -m.bEbola virus complete ppt students of jcpt -m.b
Ebola virus complete ppt students of jcpt -m.b
BHASKAR43
 
Ebola
Ebola Ebola
EBOLA.pptx
EBOLA.pptxEBOLA.pptx
EBOLA.pptx
ShubhamKumarDey
 
EBOLA.pptx
EBOLA.pptxEBOLA.pptx
EBOLA.pptx
ShubhamDey36
 
EBOLA.pptx
EBOLA.pptxEBOLA.pptx
EBOLA.pptx
ShubhamDey36
 
Hepatitis C
Hepatitis C Hepatitis C
Hepatitis C
Melaku Yetbarek,MD
 
Ebola virus ppt
Ebola virus pptEbola virus ppt
Ebola virus ppt
Appy Akshay Agarwal
 
Epstein barr virus [autosaved]
Epstein barr virus [autosaved]Epstein barr virus [autosaved]
Epstein barr virus [autosaved]
Karthick Kumar
 
Marburg Virus
Marburg VirusMarburg Virus
Marburg Virus
Julie Kwhl
 
HIV/AIDS
HIV/AIDSHIV/AIDS
HIV/AIDS
Suprakash Das
 
Ebolav2
Ebolav2Ebolav2

Similar to Branche-Hors these (20)

JSResEssay15-Final-TCDkparsad.doc
JSResEssay15-Final-TCDkparsad.docJSResEssay15-Final-TCDkparsad.doc
JSResEssay15-Final-TCDkparsad.doc
 
EBOLA & OTHER VHF 2022.pptx
EBOLA & OTHER VHF 2022.pptxEBOLA & OTHER VHF 2022.pptx
EBOLA & OTHER VHF 2022.pptx
 
Ebola.pptx
Ebola.pptxEbola.pptx
Ebola.pptx
 
Ebola virus disease - A comprehensive review
Ebola virus disease - A comprehensive reviewEbola virus disease - A comprehensive review
Ebola virus disease - A comprehensive review
 
Ebola Viruse slid show.docx
Ebola Viruse slid show.docxEbola Viruse slid show.docx
Ebola Viruse slid show.docx
 
Genomic sm musa marena (1)
Genomic sm musa marena (1)Genomic sm musa marena (1)
Genomic sm musa marena (1)
 
Ebola virus a to z
Ebola virus a to zEbola virus a to z
Ebola virus a to z
 
EBOLA VIRUS
EBOLA VIRUSEBOLA VIRUS
EBOLA VIRUS
 
Analysis Of Avian Avulavirus
Analysis Of Avian AvulavirusAnalysis Of Avian Avulavirus
Analysis Of Avian Avulavirus
 
Ebola virus complete ppt students of jcpt -m.b
Ebola virus complete ppt students of jcpt -m.bEbola virus complete ppt students of jcpt -m.b
Ebola virus complete ppt students of jcpt -m.b
 
Ebola
Ebola Ebola
Ebola
 
EBOLA.pptx
EBOLA.pptxEBOLA.pptx
EBOLA.pptx
 
EBOLA.pptx
EBOLA.pptxEBOLA.pptx
EBOLA.pptx
 
EBOLA.pptx
EBOLA.pptxEBOLA.pptx
EBOLA.pptx
 
Hepatitis C
Hepatitis C Hepatitis C
Hepatitis C
 
Ebola virus ppt
Ebola virus pptEbola virus ppt
Ebola virus ppt
 
Epstein barr virus [autosaved]
Epstein barr virus [autosaved]Epstein barr virus [autosaved]
Epstein barr virus [autosaved]
 
Marburg Virus
Marburg VirusMarburg Virus
Marburg Virus
 
HIV/AIDS
HIV/AIDSHIV/AIDS
HIV/AIDS
 
Ebolav2
Ebolav2Ebolav2
Ebolav2
 

Branche-Hors these

  • 1. Examen écrit Université de Genève Faculté de Médecine Ebola, an update Emilie BRANCHE Directeur de thèse: Prof. Francesco NEGRO Co-directrice de thèse: Dr. Sophie CLÉMENT Examinateurs: Prof. Laurent KAISER Dr. Glauciá PARANHOS-BACCALA 2015
  • 2. 2 Abstract The current outbreak of Ebola principally affecting Guinea, Sierra Leone and Liberia has caused at least 24 000 infections and over 9 000 deaths as of March 2015. The "imported" cases in Spain, United Kingdom and United States of America lead to an important surge of media, medical and research interest. This review summarizes the current knowledge about Ebola virus including the epidemiology, the pathogenesis as well as the treatments both currently available and in development. We will see that Ebola pathogenesis is definitely complex and that both coagulation and immune response play a crucial role in its etiology. I - Introduction 1- Classification Ebola virus belongs to the Ebolavirus genus in the Filoviridae family in the Mononegavirales order. Filoviridae family includes Marburgvirus with Marburgvirus, Cuevavirus with Lloviu cuevavirus and Ebolavirus. Since the original description in 1976 of the Zaire Ebolavirus , four species have emerged, Bundibugyo, Cote d'Ivoire or Tai Forest, Reston and Sudan ebolavirus [1] (Figure 1). Figure 1 : Taxonomy of the Filoviridae family adapted from Kuhn JH et al, 2010 [2]
  • 3. 3 All these species are pathogenic for humans, except Reston ebolavirus, which is pathogenic for non-human primates. These viruses have been sequenced and their molecular evolution described [3]. The current circulating Ebolavirus (EBOV) shares 97% homology with Zaire Ebolavirus [4]. This review will mainly focus on EBOV. 2 - Epidemiology The first Ebola cases appeared in 1976 in Sudan [5] and in Zaire [6]. In southern Sudan, 284 cases appeared causing 151 deaths (53% of death) in 6 months (June to November). In northern Zaire (near the Ebola river giving the name to this virus), 318 cases were described leading to 280 deaths (88% of death) in 2 months (September and October). Since these original cases, approximately 20 additional outbreaks occurred between 1976 and 2013, causing nearly 2500 deaths in the Democratic Republic of Congo, Sudan, Gabon, Ivory Coast, Uganda and Zaire [7, 8]. Since December 2013, a new outbreak is occurring in several African countries (Mali, Senegal, Nigeria, Guinea, Sierra Leone and Liberia). One case has been described in United Kingdom, another in Spain and 4 cases in the United States of America. This outbreak caused 24 282 infections so far with a mortality rate of 40% [9] (Figure 2 and Figure S1). Figure 2 : 8 March 2015: situation of EBOV outbreak.
  • 4. 4 a - Reservoir Although the primary animal host for EBOV is still unclear, fruit bats seem to be its reservoir. After several outbreaks in Gabon and Zaire between 2001 and 2005 that devastated local gorilla and chimpanzee populations, a team of researchers captured 1030 animals including bats, birds and small terrestrial vertebrates close to infected gorilla and chimpanzee carcasses. They could detect immunoglobulin G (IgG) specific for EBOV virus in sera from three different bat species, Hypsignathus monstrosus, Epomops franqueti and Myonycteris torquata. Moreover, viral genome was detected in the same bat populations in organs known to be the principal targets of EBOV, namely the liver and the spleen [10]. However, many questions regarding the mechanism by which bats are infected and transmit the viruses remain unanswered. Importantly, the transmission is not only due to direct contact between human or non-human primates and living or dead bats. Indeed the majority of new infections during outbreaks were due to human-human contacts through blood, secretions or body fluids including sweat, saliva and tears [11]. b - Clinical characteristics During the early stages, EBOV infection triggers several symptoms such as fever, severe headache, muscle pain, intense weakness, fatigue, diarrhea, vomiting and abdominal (stomach) pain. During the intermediate or advanced stages, inflammatory factors-induced vasodilatation results in both internal and external hemorrhages (bleeding or bruising). In addition to coagulation system disorders, the infection of kidney and liver leads to organ dysfunctions. Body injury and viral spread in blood circulation and organs lead to a vicious downward spiral. If viral spread cannot be controlled, patients may succumb to organ failure or secondary bacterial infection. Hemorrhage observed during EBOV disease is due to disseminated intravascular coagulation (DIC) development. This pathology is characterized by a widespread activation of clotting cascade resulting in blood clots formation in blood vessels, impairing the tissue blood flow and leading to ischemia and organ damages. In addition, this blood clot formation exhausts the coagulation factors, preventing normal coagulation and leading to hemorrhages. The mechanisms leading to DIC will be further detailed below (see chapter III.2). Furthermore, there is a weak inflammatory response coupled with a significant lymphoid cell apoptosis leading to lymphopenia, which seems to be a marker of poor prognosis. These symptoms may appear anywhere from 2 to 21 days after EBOV exposure, but the average is between 8-10 days. In most cases, the cause of death is mainly due to organ failure (such as liver or spleen) rather than to hemorrhagic fever. A major complication for the EBOV diagnostic is that symptoms observed during the early stages of infection are non-specific and difficult to distinguish from other endemic diseases, such as Lassa fever, malaria, cholera or typhoid fever.
  • 5. 5 II - The virus 1 - Morphology and genome organization Viral particles have a filamentous morphology. The origin of the name of Filoviridae family comes from the Latin word filum referring to this particular morphology (Figure 3a). EBOV is an enveloped, single strand, non-segmented, negative sense RNA virus. The 19kb viral genome contains seven genes separated by regulatory regions composed of the 3' nontranslated region (NTR), highly conserved transcription stop and start signals and the 5'NTR. The conserved transcription stop and start signal either overlap or are separated by intergenic regions (IGR) [12]. Even if the viral genome contains only seven genes, more proteins are produced through cotranscriptional editing of the GP (glycoprotein) gene [13]. Encoded proteins are nucleoprotein (NP), polymerase cofactor VP35, matrix protein VP40, glycoprotein (GP), transcriptional activator VP30, second matrix protein VP24 and RNA- dependent-RNA polymerase (L) proteins. In addition, through RNA editing EBOV is able to express two truncated secreted proteins, glycoprotein (sGP) and small glycoprotein (ssGP) [13] (Figure 3b). The viral RNA is encapsidated by NP and associated to VP35, VP30 and L to form the ribonucleoprotein (RNP) complex. The RNP is surrounded by a matrix structure, containing the matrix proteins VP40 and VP24, and finally by a host cell-derived membrane in which the surface glycoprotein GP is embedded [14]. GP self-associates as a trimer, linked by a single disulfide bond to form spikes at the virion surface [15] (Figure 3c). In addition to their structural function, these proteins play several roles notably in immune system evasion as described below (see the chapter V) Figure 3 : EBOV morphology observed by electron microscopy (a) (CDC, 2005) and schematic organization of genome (b) [14] and virion (c) [16]
  • 6. 6 2 - EBOV protein functions a - RNP complex NP, VP35, VP30 and L proteins play a fundamental role in RNP complex formation and in viral transcription and replication. NP is a 739 amino-acids (aa) protein encoded by the first gene located at the 3' region of the genome. It plays a central role in virus replication, NP together with VP24 and VP35 are necessary and sufficient for the formation of nucleocapsids that are morphologically indistinguishable from those from EBOV infected cells [17]. VP35 protein is composed of 321 aa (35kDa). In addition to its role in nucleocapsid formation by creating a link between L and N, VP35 is also a cofactor of the RNA-dependent RNA polymerase complex. It plays an important role in antiviral and IFN response inhibition detailed later (see chapter V.2). VP30 (288aa, 32kDa) interacts also with NP in the RNP complex. VP30 is a transciptional activator. VP30 can switch from a phosphorylated inactive state to an active state, through dephosphorylation by the cellular protein phosphatase 1 (PP1). This regulation maintains the balance between transcription and replication, as VP30 activity is required for the transcription initiation [18]. When VP30 is active, transcription and protein synthesis occur, while when it is inactive, viral replication takes place. The L gene encodes a large protein of 2212 aa (252kDa), highly conserved across Ebola species. This RNA-dependent-RNA polymerase (similar to the other polymerases of negative single stranded RNA virus) is responsible for the viral transcription as well as for the RNA replication. Moreover, it regulates the GP editing leading to the generation of sGP and ssGP. b - Matrix proteins: VP40 and VP24 VP40 is composed of 326 aa (35kDa) and is the most conserved and the most abundant protein in the virion. It is not clear whether the majority of VP40 in the cytoplasm or premembrane zone is monomeric [19-22] or dimeric [23] or both [24]. This VP40 form was found to be critical for both the transport of the nucleocapsid to the cell surface and for its incorporation into virions [23]. Nevertheless, this monomeric or dimeric conformation can be switched into either octameric or hexameric structures that have distinct functions. Octamer formation is critically dependent on RNA binding [25], as no octamer can be observed in the absence of RNA [26], suggesting that they may play an important role in EBOV transcription and replication [20]. Hexamers are believed to be induced by the VP40 binding to plasma membrane [19, 27] and may be implicated in the initiation of virus assembly, binding and budding via their interaction with the cytoplasmic tails of viral GP and/or the RNP complex [28, 29]. Hexameric VP40 induces host cell membrane curvature
  • 7. 7 needed for viral egress [24, 30]. This matrix protein plays a central role in the formation of the filamentous structure of EBOV virions [23, 29]. However, how VP40 induces the formation of the particular filamentous morphology of the particle is mostly unknown. In addition, a soluble secreted form of VP40 was observed during EBOV infection in vitro and was also found in the serum of virus-infected animals albeit in low amounts [31]. The role of this soluble form of VP40 as well as the mechanism by which it is released are unknown. Nevertheless, the early appearance of anti-VP40 antibodies in EBOV infected patients could be explained by the presence of this secreted VP40 [32, 33]. These observations suggest that soluble VP40 may play a role in EBOV pathogenicity. VP24, composed of 251 aa (28kDa), plays a structural role of matrix but has also a function during EBOV life cycle. In contrast to what has been reported in previous studies, Watt A et al (2014) demonstrated that VP24 has only a very modest influence on genome replication and transcription. Nevertheless, it plays an important role in particle infectivity due to its function in nucleocapsid assembly and more specifically in RNA incorporation into viral particles [34]. Like VP35, VP24 interferes with IFN response (see below in chapter V.2). c - GP The GP gene of EBOV contains an editing site allowing the translation of three differents proteins (Figure 4). The first isoform is a structural protein, translated into a glycoprotein precursor (GP0) further cleaved by a cellular proprotein convertase furin [35]. This produces a surface subunit GP1 and a transmembrane subunit GP2 that are able to form a heterotrimer. GP plays a role in virion attachment and fusion but this process remains poorly understood. GP1 contains an excessively O-linked glycosylated mucin-like region (MLR) at C- terminal, a heavily N-linked glycosylated glycan cap domain (GCD) and a receptor binding domain (RBD) which mediate the binding to a variety of host cell surface factors including T- cell immunoglobulin and mucin domain 1 (Tim-1) [36]. MLR is required for neither the viral entry nor the cellular tropism [15], as MLR-deleted GP is able to mediate viral attachment and entry, but it may influence the EBOV capacity to escape the immune system [37]. GP2 with the fusion peptide is required for the virus-host membrane fusion. In addition to this transmembrane GP form, several soluble GPs have been described. A trimeric soluble GP, called shed GP, is produced by the release of virion-attached GP byTNF-α-converting enzyme (TACE) through a cleavage site proximal to the transmembrane anchor. Moreover, GP gene encodes two non-structural forms of GP that are soluble and secreted in important quantity by infected cells. The soluble GP (sGP) is homodimeric whereas the small soluble GP (ssGP) is monomeric. During EBOV infection, the ratio between sGP and GP transcripts is approximately 75% / 20% and ssGP represents 5% of GP transcripts [38]. These secreted GPs are easily detectable in the blood of infected patients [39] and play several roles in both cytoxicity induced by EBOV and immune evasion as detailed later (see chapter III.3 and
  • 8. 8 V.1.b). In addition, a study demonstrated that sGP can substitute GP1 to form sGP-GP2 complex, suggesting a role for sGP as a structural protein [40]. Figure 4 : Processing of EBOV glycoproteins from Cook et al, 2013 [41] 3 - Viral life cycle EBOV life cycle is similar to life cycles of other viruses with negative single strand RNA (Figure 5). After GP binding to attachment factors (including DC-SIGN, L-SIGN) [42] and entry receptors, such as Tim-1 [43, 44], whole virions are internalized via macropinocytosis and trafficked to the endosomal compartment [45, 46]. GP1 is then cleaved by the endosomal cysteine proteases cathepsin B (CatB) and L (CatL) that remove the hyper-glycosylated region, which exposes the RBD in order to bind the Niemann-Pick C1 (NPC1) cholesterol transporter. GP1-NPC1 interaction leads to conformation change of trimeric GPs and allows the insertion of three fusion peptides located at the N-terminal region of GP2 in endosomal membrane. This step is essential for the fusion process, allowing viral genome release into the cytoplasm [47, 48]. The released viral RNA is then first transcribed. Due to the presence of transcription stop and start signal in the regulatory region between each gene, the negative-strand RNA genome is transcribed by the L polymerase into seven monocistronic mRNAs. These mRNAs are capped and polyadenylated. It is believed that for EBOV, such as for all negative RNA viruses, the polymerase accesses to the viral genes via a single
  • 9. 9 polymerase binding site at the 3' end. Once bound the viral polymerase progresses along the RNA template by stopping and reinitiating at each gene junction and transcribes genes in a sequential and gradient manner. Accordingly the first gene, NP, is transcribed at the highest level whereas the last gene, L is transcribed at the lowest level. Then, replication likely begins when enough NP is present to encapsidate neo-synthetized antigenome and genomes. GP-encoding mRNAs transit to the endoplasmic reticulum (ER) where GP is synthesized and form trimers. After the addition of N and O-linked glycans in the ER and Golgi apparatus, GPs are delivered to the plasma membrane by secretory vesicles. NP, VP35 and VP30 proteins associate with viral RNA to form RNP complex, and with matrix proteins (VP40 and VP24) and GP proteins. Eventually, viral particles bud at the cell surface and are released. Figure 5 : EBOV life cycle, from White JM 2012 A new player in the puzzle of filovirus entry [49]
  • 10. 10 III - Pathogenesis 1 - Target cells and tissues The detailed pathogenesis of the disease is not well understood. Nevertheless, it has been found that EBOV has a broad cell tropism, infecting a wide range of cell types. In situ hybridization and electron microscopy analyses of tissues from patients with fatal disease or from experimentally infected non-human primates showed that monocytes, macrophages, dendritic cells (DCs), endothelial cells, fibroblasts and several types of epithelial cells such as hepatocytes and adrenal cortical cells support EBOV replication [50-54]. Temporal in vivo studies in non-human primates experimentally infected with EBOV determined that monocytes, macrophages, DCs but also natural killer (NK) cells are the first and favorite targets of the virus, whereas all others cells cited above are infected much later during the course of the disease, proximal to death [51, 52, 55]. Monocytes, macrophages, and DCs appear to play a major role in the dissemination of the virus. Immunohistochemical studies have shown that the virus disseminates from lymph nodes via lymphatic and vascular systems to several organs including liver, spleen, lung, kidney, pancreas, large and small intestines and skin amongst others [50, 54]. Nevertheless, the most prominent damages are observed in liver and spleen. In these organs, cell necrosis and apoptosis were detected. The same was observed in lymph nodes leading to the lymphoid depletion detailed below see chapter IV.2). In the liver, hepatocytes and Kupffer cells are infected, leading to hepatic dysfunction directly resulting from viral damages or circulatory impairment. EBOV infection leads to coagulopathy through damages to both liver, which is the production site of clotting factors, as well as certain coagulation inhibitors, [56] and endothelial cells, which provide tissue factor (TF also known as thromboplastin), tissue factor pathway inhibitor (TFPI) and receptor for protein C activation [50, 57]. These organ disorders contribute more to the patient death than the hemorrhagic fever. 2 - Coagulation anomalies and vascular endothelium impact Coagulopathy has been observed during EBOV infection and might have several causes including activation of cytokine secretion, platelet aggregation and consumption, activation of the coagulation cascade, deficiency of coagulation factors due to both liver and endothelium damages. Indeed, it has been described that pro-inflammatory cytokines such as IL-6 are increased in human and non-human primates infected by EBOV [58, 59]. IL-6 is known to trigger the coagulation cascade. Accordingly, the transcriptional targets of IL-6 including several proteins that either increase the transcription of pro-coagulant proteins like TF or decrease the transcription of anticoagulant proteins such as antithrombin [60]. Moreover, EBOV infected monocytes and macrophages induce an increase of TF protein level in macaques circulation [61]. EBOV infection also causes hepatic necrosis and apoptosis leading to an impairment of the synthesis of critical coagulation factor including protein C,
  • 11. 11 protein S and fibrinogen [62, 63]. Deregulation of this coagulation pathway leads to disseminated intravascular coagulation (DIC) which is observed during infection and likely contributes to hemorrhage symptoms and multi-organ failure [6, 61, 64]. In addition to these problems in the coagulation pathway, the widespread injury to endothelial cells via a direct cytotoxic effect of GP (detailed later in chapter III.3) is observed in EBOV infection and is another mechanism triggering DIC. These cells have several properties, one of these being the capacity to regulate the process of coagulation and fibrinolysis and to modulate the fibrin deposition. At steady state, endothelial cell surface is thought to be essentially anticoagulant or non-thrombogenic. The control of coagulation is exerted by endothelial cells at different critical steps of the clotting cascade. Briefly, endothelial cells are the main source of TFPI, which blocks TF, the principal initiator of the coagulation cascade [65, 66]. TF is a transmembrane glycoprotein receptor expressed in response to injury at the surface of a variety of cells, including platelets, monocytes, macrophages, fibroblasts, and endothelial cells [67]. In addition, endothelial cells express a large amount of heparan sulfate and related glycosaminoglycans to neutralized clotting enzymes such as factor Xa and thrombin [65]. Eventually, endothelial cells play a critical role in the protein C anticoagulant pathway by deregulating its expression [68]. EBOV infection leads to impairment of the endothelial barrier integrity and to an increased endothelial permeability [51]. In addition, several factors secreted by both infected monocytes and macrophages can exert changes in the vascular endothelium in a variety of ways. This includes either an indirect induction of endothelial cell activation, by infecting and activating leukocytes and triggering the synthesis and local production of pro-inflammatory soluble factors, or a direct induction of changes in endothelial cell expression of cytokines, chemokines and cell adhesion molecules in the absence of immune mediators (as a direct result of virus infection, mechanism detailed in chapter III.3). Mediators released from EBOV-activated endothelial cells can modulate vascular tone, thrombosis, and/or inflammation including nitric oxide (NO), prostacyclin, interferons (IFNs), interleukin (IL)-1, IL-6, and chemokines such as IL-8, IL-6, IL-7 [61]. All these endothelial cells impairments are implicated in DIC syndrome and hemorrhage development. However, as previously mentioned, the hemorrhage observed during EBOV infection is insufficient to cause the death, as the massive loss of blood is atypical and, when is present, is largely restricted to the gastrointestinal tract. Nevertheless, it seems that pro- inflammatory cytokines secreted by monocytes, macrophages or DCs and both apoptosis and necrosis observed in several organs including liver and spleen induced by EBOV infection might participate to malfunction of both vascular system and coagulation, leading to general failure of several organs, septic shock and death.
  • 12. 12 3 - Direct toxicity In vitro studies have shown that GP has direct cytotoxic properties on endothelial cells via morphology changes leading to cell rounding and detachment [69, 70]. Indeed, studies identified a reduction at the cell surface of the expression of adhesion molecules such as integrins or immune molecules (including major histocompatibility complex class I [MHC] and the epidermal growth factor receptor) induced by GP expression. This is believed to contribute to the cell rounding and consequent loss of cell adhesion observed in infected cells [70-74]. This finding suggests that GP and more particularly the MLR of GP plays an important role in endothelial cell toxicity and could be responsible for both endothelial integrity disruption and increased endothelial permeability, triggering hemorrhage development during the disease [69]. The mechanism by which GP has this toxic effect has been shown to be dependent on GTPase dynamin. Through its interaction with dynamin, GP disrupts the normal intracellular trafficking of the cell surface proteins essential for cell attachment and immune signaling [70]. Nevertheless, the importance of GP cytotoxicity in viral pathogenesis is however controversial. Indeed, direct damages to the endothelial cells by virus replication have been observed only in animal models at terminal stages of the disease [51]. A study demonstrated that moderate expression of GP (similar to the amount observed in EBOV infected cells during the early stages of infection) did not result in morphological changes and was not cytotoxic, suggesting that cell rounding and downregulation of the surface markers are late events in EBOV infection, whereas production and massive release of virus particles occur at early steps [75]. It has been described that EBOV-infected cells release proteolytic endosomal enzymes, such as the cathepsin proteases implicated in extracellular matrix degradation and disease progression [76, 77]. The secretion of cathepsins by EBOV-infected cells suggests that these molecules may be implicated in direct cytotoxicity induced by EBOV and contribute to the vascular endothelium destruction because these proteases in vitro catalyze the degradation of extracellular matrix and induce cell rounding and detachment in vitro. A recent study showed that GP increases the NK cell toxicity. In fact, mouse macrophages infected with VSV particles containing EBOV-GP instead of their glycoprotein (VSVΔG/EBOV- GP) particles causes an increase in NK cell cytotoxicity through a decrease of MHC-I expression [55].
  • 13. 13 IV - Immune response during EBOV infection Several studies have shown that EBOV infection was associated with aberrant innate immune responses and with global suppression of adaptive immunity (Figure 6). 1 - Innate response After the epithelial barrier, innate immunity is defined as the first line of defense against pathogenic microbial exposure. Innate immune responses are not specific to a particular pathogen in contrast to the adaptive immune responses. Innate immune responses involve several pathways in order to distinguish self from non-self. The recognition of non-self leads to the activation of several cells, such as monocytes, macrophages, granulocytes, DCs, natural killer (NK) cells but also to the complement activation (soluble factors) followed by the cytokines production such as interferon (IFN). The interferon system represents a major innate defense against infections by viruses and other pathogens. Three classes of IFNs have been described. Type I IFNs, comprising IFN-α and IFN-β, are produced by many cell types. Type II IFNs, with IFN-γ, are generated by activated T cells and NK cells. Type III IFNs, including IFN λ1–3, are incompletely characterized, but are believed to mediate an antiviral response as well. The IFN response begins with the recognition of diverse pathogen- associated molecular patterns (PAMPs) by pattern recognition receptors (PRRs). Viruses contain several PAMPs recognized by specific PRRs. Double strand RNA (dsRNA), single strand RNA, CpG-DNA, 5'-triphosphate RNA or single strand DNA which are recognized by Toll-like receptors (TLR)-3 , TLR 7/8, TLR9, retinoic acid-inducible gene I (RIG-I) or single stranded DNA melanoma differentiation associated gene 5 (MDA-5). EBOV, being a negative strand RNA, induces IFN signaling through TLR-3, TLR-7/8 and RIG-I. The receptors can be localized in the cytoplasm, like RIG-I and MDA-5, or in membranes, like TLRs. Receptor activation leads to IFN production via IFN regulatory factor (IRF)-3 and IRF-7. This secreted IFN binds to its receptor composed of two subunits, IFN α receptor 1 (IFNAR1) and IFNAR2 at the cell surface in order to activate JAK-STAT pathway. This signaling pathway leads to the phosphorylation and subsequent dimerization of the transcription factor STAT, allowing its shuttle into the nucleus to induce transcription of interferon-stimulated genes (ISG)[78]. IFN response leads to an antiviral state but we will see later that several EBOV proteins can interfere at several levels with the JAK/STAT pathway (see chapter V.2). Altogether, these mechanisms are often sufficient to counter invading viruses. In addition, when they fail to do so, they favor the generation of host mediated humoral and cellular immune responses that limit and in most cases eliminate the invading pathogen. Several viruses, however, such as EBOV, have developed a variety of mechanisms to escape the innate immune system (detailed below). EBOV infection targets antigen-presenting cells (APC) during the early stages of infection. Since these cells play a critical role in immune responses, their infection by EBOV has dramatic consequences, notably by preventing their maturation. Indeed, in
  • 14. 14 vitro studies demonstrated that EBOV-infected DCs do not express the DC maturation/activation markers such as CD80, CD86, CD40, CD83 and MHC of class I and II needed to CD4+ and CD8+ T-cell co-simulation and activation [79, 80]. In addition, EBOV infection prevents cytokine and chemokine production implicated in inflammation regulation and immune response such as IFN-α, IFN-β, tumor necrosis factor (TNF) -α, IL-1β, IL-10, IL-6, IL-2, IL-8, IL-12 [79, 81]. During viral infection, NK cells quickly respond by triggering exocytosis of perforin and granzymes and secretion of IFN-γ, respectively mediating the destruction of infected cells or the macrophage activation. NK cells activation requires several signaling molecules including IL-12 (for the cytokines production), IFN-α and IFN-β (for the development of cytotoxic effector function) secreted by mature/activated DCs. Since EBOV infection prevents DC maturation/activation, NK cells activation is decreased [82] , which further favors virus replication. Therefore, a proper activation of NK cells could be critical for the protection of EBOV infection [83]. In such a dysregulated immune response context, it has been observed that despite a high viral load and necrotic lesions in fatal EBOV cases, only a minimal inflammation is observed in infected organs and tissues [50], probably due to a weak immune system activation. Yet, and in contrast to the negative impact of EBOV infection on DCs and NK cells, the infection of monocytes and macrophages by EBOV leads to an important secretion of pro-inflammatory cytokines such as IL-1β, IL-6, IL-8, IL-15, IL-16, TNF-α but not IFN-α and chemokines such as macrophage inflammatory protein (MIP)- 1α, MIP-β [84-86]. All these disturbances in immune cell activation and pro- and anti-inflammatory cytokines production contribute to facilitate the uncontrolled viral replication observed during EBOV infection. Indeed, it has been shown that the early innate response correlates with the survival of EBOV-infected patients. Therefore, the rapid initiation of innate response may limit EBOV infection and could be a critical condition to host survival [32]. 2 - Adaptive response Adaptive response is the third line of defense, after epithelial barrier and innate response. It is triggered after a few days of infection and is more powerful than innate immunity in combating the infection. In contrast with innate system, adaptive system develops a specific response to the antigen and allows establishing an immune memory. Briefly, after DC maturation and activation by pathogen detection, DCs migrate to lymph nodes to present antigens on their surface via MHC-I or II and express co-stimulatory factors (CD80, CD84, CD40) in order to activate T lymphocytes (CD4+ and CD8+). When pathogen-specific T cells are activated, they proliferate, leave the lymph node and migrate to infected tissues. CD8+ T cells directly kill the infected cells through their cytotoxic activity and CD4+ T cells (Th1) activate macrophages via both TCR-MHC-II interaction and cytokine production. Another CD4+ T cells (Th2) population remains in the lymph node and stimulates the proliferation and differentiation of pathogen-specific B cells through both MHC-II presentation and
  • 15. 15 cytokine production in order to promote the antigen-specific antibody production or proliferation of memory B cells. We have seen that EBOV replicates efficiently in DCs without cytokine and chemokine production and without inducing their maturation/activation. This lack of DC activation most likely results into poor immune responses by NK as seen before but also into weak T and B cell activation. In addition, fatal cases of EBOV infection are associated with a lack of detectable adaptive immunity. It has been observed that EBOV infection induces a substantial lymphopenia due to CD4+ and CD8+ T cell depletion and necrosis observed at least in spleen, thymus and lymph nodes of non survivors compared to survivors; the same was observed in experimentally infected non-human primates [50, 87, 88], and the different mechanisms implicated in this phenomenon will be described below (paragraph "lymphopenia"). Nevertheless, despite significant lymphocyte apoptosis, it has been demonstrated that a functional and specific, albeit insufficient, adaptive immune response is present in lethal EBOV infection [89], occurring even in the presence of incompletely activated DCs. There is an increased percentage of CD4+ and CD8+ T cells expressing high levels of CD44, a T-cell activation and maturation marker, close to the end of lethal EBOV infection. CD8+ T cells play an important role in EBOV infection. Indeed, in lethal mice model of EBOV, the IFN-γ production by CD8+ T cells in response to EBOV infection was observed at the end of the disease [89]. In addition, this important source of IFN-γ could explain the macrophage and monocyte activation observed during EBOV infection. Moreover, transfer of EBOV-specific CD8+ T cells from mice infected with EBOV during 7 days protects naive mice from EBOV challenge. [89]. Together, these data support the hypothesis that functional adaptive immune responses are present, at the end of the disease in lethal EBOV-infected mice but is insufficient in part due to massive lymphocyte apoptosis. Concerning B cells, a clinical study performed during the 1996 outbreak in Gabon described humoral immune responses in EBOV infected patients, as antibodies directed against GP have been found in surviving patients [90]. In addition, important levels of IgG and IgM, specific to NP, VP40 and VP35, have been found by ELISA in all survivors early in disease or during early convalescence. In contrast, no viral antigen-specific IgG have been found in fatal cases and only weak IgM levels have been detected in one-third of fatal cases [33]. These results suggest that a prompt and vigorous humoral response may help survivors to limit and finally control viral dissemination. Furthermore, it has been observed that this immunoglobulin deficiency is not associated with a decrease of B cells. The mechanism by which EBOV impacts on immunoglobulin levels therefore remains poorly understood [33, 91]. Nevertheless, the impact of EBOV infection on T cell activation and proliferation could alter B cell activation.
  • 16. 16 Lymphopenia The mechanism by which EBOV induces a lymphopenia is not fully understood, likely in part because a direct mechanism cannot be involved since EBOV does not infect lymphocytes. As discussed above, EBOV readily infects and replicates in DCs, interfering with their activation/maturation and therefore with their ability to initiate the adaptive immune response and the associated lymphocytes expansion [61, 80]. We have seen before that the release of NO, which is a physiological vasodilator and anti- platelet factor, was increased by the endothelial cells activated by EBOV [61, 92]. In vivo study demonstrated that blood levels of NO were much higher in fatal cases (increasing with disease severity), and extremely elevated levels could have negatively affected vascular tone and contributed to virus-induced shock [93]. In addition of its role in endothelial barrier, NO could also play a role in lymphopenia. Briefly, it has been shown that NO promotes apoptotic pathways in numerous cell types including lymphocytes through the indirect activation of caspases [94], moreover NO inhibits T and B cell proliferation via the downregulation of MHC-II, co-stimulation molecules and/or cytokines (such as IL-12) [95]. The death receptor pathway activation could be implicated in lymphocyte apoptosis observed during EBOV infection. EBOV infection could induce both intrinsic (mitochondrial mediated pathway) and extrinsic (death receptor pathway) cell death cascades as crosstalk occurs between these two pathways. In the intrinsic pathway, intracellular stress factors (such as oxidative stress or DNA injury) via Bax and Bak proteins cause depolarization of the mitochondrial membrane, thereby inducing the release of cytochrome C and activating the caspase cascade beginning with caspase-9. Bcl-2 protein is known to inhibit apoptosis via its interaction with Bax and Bak proteins [96]. In fatal EBOV cases, a decrease of Bcl-2 mRNA level has been observed in PBMC during the disease, whereas a strong increase has been detected in survivors at the time of T-cell activation [33]. The extrinsic pathway is initiated by ligand-receptor interaction at the cell surface including either Fas Ligand with Fas or TRAIL with TRAIL receptors (such as DR4 and DR5). Such interactions lead to caspase (caspase 8 and then caspases 3 and 7) activation via the adaptor protein Fas-associated death domain (FADD) and finally induce DNA degradation and cell death [97]. With regards to the extrinsic pathway, EBOV infection increases TRAIL expression in cultured monocyte-like cells, and some EBOV-infected monkeys exhibit an increase of soluble Fas in their sera [86]. Furthermore, TRAIL and Fas mRNA expressions are increased in the PBMC of infected monkeys [52]. Another explanation for lymphopenia induced by EBOV implicates GP. Indeed GP contains a domain with a significant homology with the "immunosuppressive peptide" found in glycoproteins of various oncogenic retroviruses known to often induce immunosuppression [98]. This will be detailed below (see chapter V.1.c). In addition, it was postulated that sGP
  • 17. 17 could play a role in lymphocyte apoptosis by interacting with circulating lymphocytes, as it was detected in large amounts in the blood [99]. Nevertheless, an in vivo study showed that sGP was not able to induce T cell apoptosis neither by itself nor by death receptor co- stimulation; further studies are required to investigate the ability of sGP to induce apoptosis via the intrinsic pathway [100]. Altogether, immune system dysfunctions (weak innate and adaptive immune system activation or lymphopenia) contribute to the uncontrolled spread and growth of the virus. This suggests that a strong immune response may result in protection against EBOV infection, which may guide the design of new therapeutic strategies to control lethal EBOV disease. Figure 6 : Model of EBOV pathogenesis in primates. Adapted to Bray M 2005 [101]
  • 18. 18 V - Immune response evasion by EBOV viral proteins Several mechanisms by which EBOV escapes immune system have been suggested (Figure 7). Figure 7 : Potential mechanisms by which various EBOV proteins evade host innate and acquired immune systems. Adapted from Ansari AA 2014 [102] 1 - GP implications a - Glycosylation and MLR EBOV infection elicits only low level of neutralizing antibodies against GP in humans and other animals [14]. As mentioned above, the heavy glycosylation of GP is implicated in the immune system escape. These glycans located in the MLR sequence promote the generation of antibodies against the more variable GP1 domain, which are not able to confer a strong protection [103]. In mice, removal of the MLR of GP1 can lead to the production of more efficient antibodies directed against the conserved glycoprotein core structure, confirming the impact of this MLR domain in masking neutralizing epitopes [104]. Moreover, O- and N- glycosylations impedes the recognition of GP by neutralizing antibodies through steric shielding [41, 74, 105-107]. In addition, MLR is necessary and sufficient to decrease the expression of cell surface proteins such as MHC-I and several members of the integrin family. This domain blocks the access to MHC-I needed for CD8+ T cell stimulation [69, 105]. An additional mechanism by which glycosylations play an important function in the immune escape involves N-linked GP2 glycosylation. Indeed, mutation of one of the two N-linked GP2 glycosylation sites prevents the interaction between GP1 and GP2 required for GP localization at the plasma membrane and is implicated in antigenicity and immunogenicity of EBOV GP. All these results suggest that it might be possible to enhance immunity by specific modifications in the GP glycosylation [37].
  • 19. 19 In addition, we have seen that EBOV infection leads to DC maturation defects and consequently to a failure of efficient T cell activation [79, 80]. In vitro studies using EBOV- virus like particles (VLP) containing VP40 demonstrated that VLPs, contrary to EBOV infection, have the capacity to activate DCs. MLR is the domain required for DC activation via a recognition of MLR by toll like receptor (TLR)-4 and NF-kappaB and MAPK signaling pathway activation [108, 109]. Indeed, VLPs with wild-type GP but not with MLR-deleted GP can activate TLR-4-dependent responses. In EBOV infection context, these results suggest that MLR plays a major role in the abnormal DC activation observed during the disease. b - Role of sGP and shed GP Secreted GPs, sGP and shed GP, have been shown to be important in immune evasion. Because sGP shares 295 amino acids with GP and is the predominant transcript for the GP gene, it has been postulated that sGP probably competes with virion-attached GP. Indeed, the majority of antibodies from EBOV-surviving patients and monkeys are directed against sGP rather than against GP1/2 [110, 111]. It is possible that the majority of antibodies binding sGP are non-neutralizing, but it is likely that the weak amount of neutralizing antibody production is absorbed by the much more abundant sGP. Indeed, it has been demonstrated that sGP serves as a decoy for neutralizing antibodies [112]. In addition to its role in adaptive response evasion, sGP has been reported to bind to neutrophils through the Fcγ receptor thereby inhibiting early neutrophil activation [113]. Concerning the shed GP, in a guinea pig model of EBOV infection, this secreted GP is present in significant amounts in the blood of infected animals. Shed GP inhibits the neutralizing activity of EBOV antibodies, and the increase of shed GP in infected animals observed between days 6 and 9 post infection correlates with the course of disease and the lethal outcome at day 9 [114]. All these findings suggest that secreted GPs may play an important role in the pathogenesis. c - Immunosuppressive domain in GP Several retroviruses including Avian reticuloendotheliosis virus (ARV) and Feline leukemia virus (FeLV), have a particular peptide in their envelope protein named p15E or "immunosuppressive peptide", that has immunosuppressive properties. For example, this peptide inhibits the T cell activation normally induced by concanavalin stimulation [115], the proliferation of murine cytotoxic T cells [116] and macrophage recruitment to the inflammatory site in mice [117]. Amino acid sequence comparison has uncovered a high homology between this "immunosuppressive peptide" and 160 residues at the C-terminal part of EBOV-GP which could explain the immunosuppressive effect mediated by EBOV-GP [98]. More recently, a study identified a 17-mer peptide in this region as the immunosuppressive domain of EBOV-GP. This peptide induces a significant decline of CD4+ and CD8+ T cells. In addition, this peptide induces a decrease of IL-2 receptor at the T cell surface, but also inhibits IFN-γ, IL-2 and IL-10 expression leading to an inhibition of T cell proliferation and activation [118]. The mechanism by which immunosuppressive peptide
  • 20. 20 acts on CD4+ and CD8+ cells is unknown but it has been hypothesized that it inactivates these cells by directly contacting them or indirectly through its previously described effect on APCs. 2 - IFN pathway inhibition by VP35 and VP24 EBOV uses several mechanisms in order to inhibit IFN production (Figure 8). It has been shown that VP35 is responsible for the absence of IFN-α production and prevents the activation of IFN-stimulated response element (ISRE)-containing promoters when either transfected dsRNA or viral infection is used as the activating stimulus [119]. A more detailed study of the mechanism by which VP35 influences the host IFN response showed that it inhibits the IFN synthesis at several levels. VP35 can bind viral dsRNA and inhibit the recognition by helicase RIG-I implicated in the IFN pathway and then the IFN-α and -β production [120]. The ability of VP35 to block IFN production was also correlated with its ability to inhibit the phosphorylation of IRF-3 through interaction with kinases including IκB kinase epsilon (IKKε) and TANK-binding kinase 1 (TBK-1) [121], and thus inhibiting its nuclear translocation and activation [122]. A SUMOylation of IRF-7 induced by VP35 was recently described as an additional mechanism of repression of the transcription of IFN genes [123]. Co-immunoprecipitation experiments demonstrated that VP35 interacts with PIAS1 (protein inhibitor of activated STAT-1) and Ubc9, two proteins involved in the small ubiquitin-like modifier (SUMO) conjugation cascade [124, 125]. Besides that, Feng et al have demonstrated that VP35 protein is a RNA binding protein with a stronger affinity for dsRNA than PKR. Consequently, VP35 competes with PKR for EBOV dsRNA binding and prevents the phosphorylation of translation initiation factor eIF-2 (eIF-2) by PKR required to stop protein synthesis and thus viral replication [126]. In addition to VP35, VP24 is another important player in the counteraction of IFN pathway by EBOV. VP24 inhibits the IFN pathway by preventing the nuclear accumulation of STAT-1 [127]. Actually, VP24 binds to karyopherin-α, a nuclear transporter, with very high affinity to compete with STAT-1 and inhibit its nuclear transport [128, 129]. In addition to the JAK-STAT pathway, the p38 mitogen-activated protein (MAP) kinase pathway is also critical for the IFN response [130]. Engagement of the IFN receptor by IFN activates a cascade of MAP kinases, leading to the phosphorylation of the alpha isoform of p38 (p38-α) [131]. Phosphorylated p38-α then triggers the phosphorylation of downstream transcription factors that participate in IFN responses. It is well established that p38 is essential for gene transcription via ISRE or GAS elements [130-132]. It has been observed that VP24 inhibits the p38 MAP kinase pathway by preventing the phosphorylation of p38-α [133]. The dual action of these two viral proteins, VP35 and VP24, may thus contribute to a potent inhibition of the IFN pathway, permitting an efficient virus replication and dissemination in the host.
  • 21. 21 Figure 8 : EBOV proteins interfering with interferon signaling
  • 22. 22 VI - Diagnosis and treatments Although EBOV is considered to be a significant public health problem, no licensed drug or vaccine is currently available [134-136]. The most effective measure for controlling disease propagation is the isolation of patients and strict barrier nursing procedures to protect healthcare workers. Meanwhile, symptomatic and supportive care is the treatment of choice. Nevertheless, owing to the advances of basic EBOV research, several promising drugs and vaccine candidates [137] are under development. 1 - Diagnosis methods As written above, the clinical symptoms in the early stages of EBOV infection are very similar to others viral diseases such as flu and other respiratory infections, common enteritis or other infections frequently occurring in African including malaria and Lassa fever. Therefore, especially in the early stages, virological testing is very important for the diagnosis. Specific EBOV-antibodies detection by ELISA and immunofluorescence has been developed but as mentioned above, EBOV antibodies are produced only in small quantity, especially in fatal cases. The inoculation of a cell culture with patient sera or other body fluids or tissue extracts is the classical method to isolate and amplify EBOV. Then, EBOV is detected by PCR or immunofluorescence using viral-specific primers or antibodies respectively. Antigen blood tests are based on the detection of virus proteins using specific antibodies and are hardly influenced by virus variability. The high viremia in EBOV patients often facilitates antigen detection, although the tests are clinically less sensitive than PCR [138, 139]. As EBOV has a specific filamentous morphology, the direct detection of EBOV by electron microscopy in organ section and serum is possible but high virus concentrations are needed [140]. A major disadvantage of these diagnosis methods is the time required to isolate the virus (days to week) and the need of biosafety level 3 or 4 facilities. The detection by electronic microscopy is not routinely used because of its high cost. Therefore, the most used method is based on nucleic acid tests, as it requires 24-48h to obtain results in a very sensitive fashion. Very recently a new test that provides results within 15 minutes has been developed, the ReEBOVTM Antigen Rapid Test. This test, which is based on the detection of the VP40 protein rather than nucleic acids, is able to correctly identify about 92% of EBOV infected patients and to exclude 85% of those not infected with the virus. In addition to its rapidity, the antigen test is easy to perform and does not require electricity, which therefore would favor its use in lower health care facilities or mobile units [141].
  • 23. 23 2 - Treatments and Vaccines Currently, the majority of treatments used aim at treating symptoms induced by EBOV. For example, as EBOV inhibits IFN signaling, exogenous INF-α or INF-β have been used and could delay the occurrence of viremia or increase survival time, but they cannot rescue non- human primates from lethal infection [142, 143]. As EBOV infection indirectly impairs the coagulation pathway by provoking the depletion of clotting factors through aberrant and excessive coagulation, the recombinant nematode anticoagulant protein c2 (rNAPc2) and the recombinant human activated protein C (rhAPC), originally used for anticoagulation purposes, have been tested and gave promising results in infected monkey [63, 144]. rNAPc2, which has shown 33% efficacy in non-human primates [144], is in Phase II trial for thrombosis prevention. Nevertheless no human trial is planned for EBOV treatment [145]. Several treatments targeting a specific step of viral life cycle including entry, RNA synthesis and translation have been developed. a - Candidates to block the viral entry In order to block the virus entry, researchers purified patients-derived polyclonal or monoclonal antibodies specifically targeting the main neutralizing epitopes on EBOV-GP. The antibody KZ52, derived from a survivor of the Kikwit EBOV outbreak in 1995, displays a potent neutralizing activity and has been shown to protect guinea pigs [146] but not non- human primates [147]. During the past years, researchers have developed three generations of antibody cocktail formulations. The first one was based on the combination of two human-mouse chimeric antibodies, ch133 and ch226, which presented strong neutralizing activity against EBOV in vitro. Unfortunately, trials in non-human primates challenged with EBOV were not convincing [148]. A second generation of anti-EBOV antibody cocktail formulas, ZMAb and MB-003 consisting of three different neutralizing antibodies derived from EBOV GP, have been tested in non-human primates. ZMAb, containing mAbs 1H3, 2G4 and 4G7, showed 100% protection in Cynomolgus macaques [149]. The MB-003 cocktail, including antibodies of c13C6, h-13F6, and c6D8, showed 67% protection in macaques [150]. It seems that human trial has started so far for this treatment. This technology may be insufficiently robust to promote the production of neutralizing antibodies to fight the current EBOV outbreak. A recent study has established a better optimized antibody combination derived Zmab and MB-003, named Zmapp and containing c13C6, 2G4 and 4G7. This new mAbs combination demonstrates a successful protection in non-human primates [151]. Phase I safety and efficacy trials have been initiated in January 2015, but the conclusions are not yet available [145]. In addition to the neutralizing antibodies, other treatments have been developed to block viral entry. Since the first C-terminal heptad repeat (CHR)-peptide-based HIV entry inhibitor
  • 24. 24 discovered in 1992 [152], this potential treatment strategy has been applied against many enveloped viruses, including EBOV [153, 154]. Briefly, as the CHR domain of GP2 plays a role during the fusion step in the endosomes, exogenous CHR could be able to compete with viral CHR and prevent the viral fusion. This treatment showed inhibition activity against three EBOV species, including Zaire, Sudan and Reston Ebolavirus [153]. Other therapeutic candidates have been described to prevent the fusion step including Cat L/B inhibitor [155] and NPC binding compounds [156]. b - Candidates to block viral RNA synthesis and/or translation Others drugs targeting RNA synthesis and translation have been developed. Nucleot(s)ides analogues including Ribavirin, Favipiravir and Brincidofovir have been tested. Ribavirin could not limit the replication of EBOV and failed to protect animals from lethal challenge [157, 158]. Interestingly, Favipiravir showed efficient antiviral activity in mouse models for EBOV infections [159]. Clinical efficacy trials began in Guinea in December 2014, however more data are required in order to draw a conclusion [145]. Brincidofovir (CMX001), showed potent anti-EBOV activity in vitro, and has been used to treat EBOV patients but its mechanism of action is unclear. However, a new phase II clinical trials of Brincidofovir has been launched to test its potential safety and antiviral activity in EBOV infected patients [160]. A new clinical efficacy trial began in Liberia in January 2015, but due to the lack of patients this trial has been stopped. In addition, to date no precise results are available because this drug is often combined with other drug therapies [145]. Finally, BCX-4430, another nucleoside analogue, interferes with the function of RNA polymerase of EBOV, and confers protection to EBOV-challenged rodent animals [161]. BCX-4430 is in phase I safety trial and efficacy trials will begin providing that the safety results from Phase I will be satisfactory [145]. Others strategies using small interfering RNAs (siRNAs) have been developed. Especially, siRNAs specifically directed against the RNA sequences of RNP complex, VP24, and VP35 were tested [162]. For instance AVI-6002, a mixture of iRNA targeting mRNA sequences of VP24 and VP35 protected five of eight rhesus monkeys from EBOV challenge [163]. For this drug, the phase I safety is completed but there are no human trial planned at this time [145]. c - Vaccines Several vaccine candidates have been tested on rodent and non-human primates [164]. The first trials were done with inactivated viruses but this method was quickly abandoned. A lot of viral vectors have been used to produced anti-EBOV vaccines including Venezuelan equine encephalitis virus [165], adenovirus [166], virus Parainfluenza [167] or Vesicular stomatitis virus (VSV) [168]. Attenuated recombinant VSV vaccine expressing EBOV GP protects non-human primates from EBOV infection. Interestingly, it has been used to successfully treat a scientist infected
  • 25. 25 by EBOV [169]. Clinical trials are in progress in several countries including United States, Canada, Germany, Gabon and Switzerland. Concerning the last one, clinical trials are performed in Geneva and began in September 2014. Initial data obtained were very promising but the development of unexpected mild to moderate joint pain 10 to 15 days after injection had lead to the suspension of this trial. In January 2015, the trial resumed using a lower dose and final results are expected soon. The appearance of reverse genetic tools for EBOV allowed the opening of a new way in the design of vaccine vectors. For example, it has been shown that EBOV recombinant carrying mutations in the domain of the VP35 involved in the suppression of IFN production loses its virulence in a guinea pig model [170]. It also effectively protects guinea pigs during EBOV infection. However, this method is unsafe since the recombinant EBOV could mutate and therby regain its pathogenic potential in the vaccinated patient. VLPs expressing immunogenic proteins such as the NP, GP and VP40 EBOV were also tested [171], but this approach is expensive and difficult to implement. VII - Conclusion This review summarizes the major knowledge on EBOV accumulated during almost 40 years. Unfortunately, the entry receptors, virus life cycle, immune response and evasion during infection are not fully understood. As of today, there are no vaccines or efficient treatment available. However, this virus has caused a lot of deaths since 1976. But the interest for the research even if it seems to correlate with death cases (Figure S2), was scanty for many years probably due to the fact that outbreaks spread only in Africa, and thus far away from Western countries. Interestingly, two cases of Marburg virus (a virus close to EBOV and with similar symptoms) have been detected in 2008 in the Netherlands [172] and the United States [173]. These cases alerted the international community on the risk of emergent viral diseases and have had a positive impact on the number of publications related to EBOV (Figure S2). In addition, the ongoing outbreak, has caused a huge increase of publications on EBOV (Figure S3). After almost 40 years and thousands of deaths, EBOV finally begins to receive some attention from researchers, and more precisely from the organizations that fund basic research and the pharmacological companies.
  • 26. 26 References 1. Kuhn, J.H., et al., Virus nomenclature below the species level: a standardized nomenclature for natural variants of viruses assigned to the family Filoviridae. Arch Virol, 2013. 158(1): p. 301-11. 2. Kuhn, J.H., et al., Proposal for a revised taxonomy of the family Filoviridae: classification, names of taxa and viruses, and virus abbreviations. Arch Virol, 2010. 155(12): p. 2083-103. 3. Carroll, S.A., et al., Molecular evolution of viruses of the family Filoviridae based on 97 whole- genome sequences. J Virol, 2013. 87(5): p. 2608-16. 4. Baize, S., et al., Emergence of Zaire Ebola virus disease in Guinea. N Engl J Med, 2014. 371(15): p. 1418-25. 5. Ebola haemorrhagic fever in Sudan, 1976. Report of a WHO/International Study Team. Bull World Health Organ, 1978. 56(2): p. 247-70. 6. Ebola haemorrhagic fever in Zaire, 1976. Bull World Health Organ, 1978. 56(2): p. 271-93. 7. WHO. Ebola virus disease, Disease outbreak news. 2014. 8. OMS. Maladie à virus Ebola. 2014. 9. WHO. Ebola Situation Report - 8 March 2015. 2015. 10. Leroy, E.M., et al., Fruit bats as reservoirs of Ebola virus. Nature, 2005. 438(7068): p. 575-6. 11. Nkoghe, D., et al., [Practical guidelines for the management of Ebola infected patients in the field]. Med Trop (Mars), 2004. 64(2): p. 199-204. 12. Neumann, G., S. Watanabe, and Y. Kawaoka, Characterization of Ebolavirus regulatory genomic regions. Virus Res, 2009. 144(1-2): p. 1-7. 13. Lee, J.E. and E.O. Saphire, Ebolavirus glycoprotein structure and mechanism of entry. Future Virol, 2009. 4(6): p. 621-635. 14. Feldmann, H.S., A. Geisbert, TW., Filoviridae: Marburg and Ebola Viruses, in Fileds Virology, D.H. Knipe, PM., Editor. 2013. p. 923-956. 15. Jeffers, S.A., D.A. Sanders, and A. Sanchez, Covalent modifications of the ebola virus glycoprotein. J Virol, 2002. 76(24): p. 12463-72. 16. Mackay, I.M. Ebola virus disease: obliterating a variant and stalled case decline... 2015. 17. Huang, Y., et al., The assembly of Ebola virus nucleocapsid requires virion-associated proteins 35 and 24 and posttranslational modification of nucleoprotein. Mol Cell, 2002. 10(2): p. 307- 16. 18. Martinez, M.J., et al., Role of Ebola virus VP30 in transcription reinitiation. J Virol, 2008. 82(24): p. 12569-73. 19. Scianimanico, S., et al., Membrane association induces a conformational change in the Ebola virus matrix protein. EMBO J, 2000. 19(24): p. 6732-41. 20. Hoenen, T., et al., VP40 octamers are essential for Ebola virus replication. J Virol, 2005. 79(3): p. 1898-905. 21. Dessen, A., et al., Crystal structure of the matrix protein VP40 from Ebola virus. EMBO J, 2000. 19(16): p. 4228-36. 22. Ruigrok, R.W., et al., Structural characterization and membrane binding properties of the matrix protein VP40 of Ebola virus. J Mol Biol, 2000. 300(1): p. 103-12. 23. Bornholdt, Z.A., et al., Structural rearrangement of ebola virus VP40 begets multiple functions in the virus life cycle. Cell, 2013. 154(4): p. 763-74. 24. Adu-Gyamfi, E., et al., Investigation of Ebola VP40 assembly and oligomerization in live cells using number and brightness analysis. Biophys J, 2012. 102(11): p. 2517-25. 25. Gomis-Ruth, F.X., et al., The matrix protein VP40 from Ebola virus octamerizes into pore-like structures with specific RNA binding properties. Structure, 2003. 11(4): p. 423-33. 26. Timmins, J., et al., Oligomerization and polymerization of the filovirus matrix protein VP40. Virology, 2003. 312(2): p. 359-68.
  • 27. 27 27. Adu-Gyamfi, E., et al., The Ebola virus matrix protein penetrates into the plasma membrane: a key step in viral protein 40 (VP40) oligomerization and viral egress. J Biol Chem, 2013. 288(8): p. 5779-89. 28. Jasenosky, L.D., et al., Ebola virus VP40-induced particle formation and association with the lipid bilayer. J Virol, 2001. 75(11): p. 5205-14. 29. Noda, T., et al., Ebola virus VP40 drives the formation of virus-like filamentous particles along with GP. J Virol, 2002. 76(10): p. 4855-65. 30. Soni, S.P., et al., The Ebola virus matrix protein deeply penetrates the plasma membrane: an important step in viral egress. Biophys J, 2013. 104(9): p. 1940-9. 31. Reynard, O., et al., Unconventional secretion of Ebola virus matrix protein VP40. J Infect Dis, 2011. 204 Suppl 3: p. S833-9. 32. Leroy, E.M., et al., Early immune responses accompanying human asymptomatic Ebola infections. Clin Exp Immunol, 2001. 124(3): p. 453-60. 33. Baize, S., et al., Defective humoral responses and extensive intravascular apoptosis are associated with fatal outcome in Ebola virus-infected patients. Nat Med, 1999. 5(4): p. 423-6. 34. Watt, A., et al., A novel life cycle modeling system for Ebola virus shows a genome length- dependent role of VP24 in virus infectivity. J Virol, 2014. 88(18): p. 10511-24. 35. Volchkov, V.E., et al., Processing of the Ebola virus glycoprotein by the proprotein convertase furin. Proc Natl Acad Sci U S A, 1998. 95(10): p. 5762-7. 36. Lee, J.E., et al., Structure of the Ebola virus glycoprotein bound to an antibody from a human survivor. Nature, 2008. 454(7201): p. 177-82. 37. Dowling, W., et al., Influences of glycosylation on antigenicity, immunogenicity, and protective efficacy of ebola virus GP DNA vaccines. J Virol, 2007. 81(4): p. 1821-37. 38. Volchkov, V.E., et al., Recovery of infectious Ebola virus from complementary DNA: RNA editing of the GP gene and viral cytotoxicity. Science, 2001. 291(5510): p. 1965-9. 39. Sanchez, A., et al., Detection and molecular characterization of Ebola viruses causing disease in human and nonhuman primates. J Infect Dis, 1999. 179 Suppl 1: p. S164-9. 40. Iwasa, A., M. Shimojima, and Y. Kawaoka, sGP serves as a structural protein in Ebola virus infection. J Infect Dis, 2011. 204 Suppl 3: p. S897-903. 41. Cook, J.D. and J.E. Lee, The secret life of viral entry glycoproteins: moonlighting in immune evasion. PLoS Pathog, 2013. 9(5): p. e1003258. 42. Matsuno, K., et al., C-type lectins do not act as functional receptors for filovirus entry into cells. Biochem Biophys Res Commun, 2010. 403(1): p. 144-8. 43. Kondratowicz, A.S., et al., T-cell immunoglobulin and mucin domain 1 (TIM-1) is a receptor for Zaire Ebolavirus and Lake Victoria Marburgvirus. Proc Natl Acad Sci U S A, 2011. 108(20): p. 8426-31. 44. Moller-Tank, S., et al., Role of the phosphatidylserine receptor TIM-1 in enveloped-virus entry. J Virol, 2013. 87(15): p. 8327-41. 45. Aleksandrowicz, P., et al., Ebola virus enters host cells by macropinocytosis and clathrin- mediated endocytosis. J Infect Dis, 2011. 204 Suppl 3: p. S957-67. 46. Gnirss, K., et al., Cathepsins B and L activate Ebola but not Marburg virus glycoproteins for efficient entry into cell lines and macrophages independent of TMPRSS2 expression. Virology, 2012. 424(1): p. 3-10. 47. Ng, M., et al., Cell entry by a novel European filovirus requires host endosomal cysteine proteases and Niemann-Pick C1. Virology, 2014. 468-470: p. 637-46. 48. Carette, J.E., et al., Ebola virus entry requires the cholesterol transporter Niemann-Pick C1. Nature, 2011. 477(7364): p. 340-3. 49. White, J.M. and K.L. Schornberg, A new player in the puzzle of filovirus entry. Nat Rev Microbiol, 2012. 10(5): p. 317-22.
  • 28. 28 50. Martines, R.B., et al., Tissue and cellular tropism, pathology and pathogenesis of Ebola and Marburg viruses. J Pathol, 2015. 235(2): p. 153-74. 51. Geisbert, T.W., et al., Pathogenesis of Ebola hemorrhagic fever in primate models: evidence that hemorrhage is not a direct effect of virus-induced cytolysis of endothelial cells. Am J Pathol, 2003. 163(6): p. 2371-82. 52. Geisbert, T.W., et al., Pathogenesis of Ebola hemorrhagic fever in cynomolgus macaques: evidence that dendritic cells are early and sustained targets of infection. Am J Pathol, 2003. 163(6): p. 2347-70. 53. Zaki, S.R., 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: p. S36-47. 54. Ryabchikova, E.I., L.V. Kolesnikova, and S.V. Luchko, An analysis of features of pathogenesis in two animal models of Ebola virus infection. J Infect Dis, 1999. 179 Suppl 1: p. S199-202. 55. Williams, K.J., et al., VSVDeltaG/EBOV GP-Induced Innate Protection Enhances Natural Killer Cell Activity to Increase Survival in a Lethal Mouse Adapted Ebola Virus Infection. Viral Immunol, 2015. 28(1): p. 51-61. 56. Blonski, W., T. Siropaides, and K.R. Reddy, Coagulopathy in liver disease. Curr Treat Options Gastroenterol, 2007. 10(6): p. 464-73. 57. van Hinsbergh, V.W., Endothelium--role in regulation of coagulation and inflammation. Semin Immunopathol, 2012. 34(1): p. 93-106. 58. Hensley, L.E. and T.W. Geisbert, The contribution of the endothelium to the development of coagulation disorders that characterize Ebola hemorrhagic fever in primates. Thromb Haemost, 2005. 94(2): p. 254-61. 59. Leroy, E.M., et al., Human asymptomatic Ebola infection and strong inflammatory response. Lancet, 2000. 355(9222): p. 2210-5. 60. Kerr, R., D. Stirling, and C.A. Ludlam, Interleukin 6 and haemostasis. Br J Haematol, 2001. 115(1): p. 3-12. 61. Geisbert, T.W., et al., Mechanisms underlying coagulation abnormalities in ebola hemorrhagic fever: overexpression of tissue factor in primate monocytes/macrophages is a key event. J Infect Dis, 2003. 188(11): p. 1618-29. 62. Ebihara, H., et al., Host response dynamics following lethal infection of rhesus macaques with Zaire ebolavirus. J Infect Dis, 2011. 204 Suppl 3: p. S991-9. 63. Hensley, L.E., et al., Recombinant human activated protein C for the postexposure treatment of Ebola hemorrhagic fever. J Infect Dis, 2007. 196 Suppl 2: p. S390-9. 64. Zampieri, C.A., N.J. Sullivan, and G.J. Nabel, Immunopathology of highly virulent pathogens: insights from Ebola virus. Nat Immunol, 2007. 8(11): p. 1159-64. 65. Bombeli, T., M. Mueller, and A. Haeberli, Anticoagulant properties of the vascular endothelium. Thromb Haemost, 1997. 77(3): p. 408-23. 66. Broze, G.J., Jr., Tissue factor pathway inhibitor. Thromb Haemost, 1995. 74(1): p. 90-3. 67. Chu, A.J., Tissue factor, blood coagulation, and beyond: an overview. Int J Inflam, 2011. 2011: p. 367284. 68. Esmon, C.T., The anticoagulant and anti-inflammatory roles of the protein C anticoagulant pathway. J Autoimmun, 2000. 15(2): p. 113-6. 69. Yang, Z.Y., et al., Identification of the Ebola virus glycoprotein as the main viral determinant of vascular cell cytotoxicity and injury. Nat Med, 2000. 6(8): p. 886-9. 70. Sullivan, N.J., et al., Ebola virus glycoprotein toxicity is mediated by a dynamin-dependent protein-trafficking pathway. J Virol, 2005. 79(1): p. 547-53. 71. Simmons, G., et al., Ebola virus glycoproteins induce global surface protein down-modulation and loss of cell adherence. J Virol, 2002. 76(5): p. 2518-28.
  • 29. 29 72. Escudero-Perez, B., et al., Shed GP of Ebola virus triggers immune activation and increased vascular permeability. PLoS Pathog, 2014. 10(11): p. e1004509. 73. Ray, R.B., et al., Ebola virus glycoprotein-mediated anoikis of primary human cardiac microvascular endothelial cells. Virology, 2004. 321(2): p. 181-8. 74. Reynard, O., et al., Ebolavirus glycoprotein GP masks both its own epitopes and the presence of cellular surface proteins. J Virol, 2009. 83(18): p. 9596-601. 75. Alazard-Dany, N., et al., Ebola virus glycoprotein GP is not cytotoxic when expressed constitutively at a moderate level. J Gen Virol, 2006. 87(Pt 5): p. 1247-57. 76. Barrientos, L.G. and P.E. Rollin, Release of cellular proteases into the acidic extracellular milieu exacerbates Ebola virus-induced cell damage. Virology, 2007. 358(1): p. 1-9. 77. Nomura, T. and N. Katunuma, Involvement of cathepsins in the invasion, metastasis and proliferation of cancer cells. J Med Invest, 2005. 52(1-2): p. 1-9. 78. Schoggins, J.W., et al., A diverse range of gene products are effectors of the type I interferon antiviral response. Nature, 2011. 472(7344): p. 481-5. 79. Bosio, C.M., et al., Ebola and Marburg viruses replicate in monocyte-derived dendritic cells without inducing the production of cytokines and full maturation. J Infect Dis, 2003. 188(11): p. 1630-8. 80. Mahanty, S., et al., Cutting edge: impairment of dendritic cells and adaptive immunity by Ebola and Lassa viruses. J Immunol, 2003. 170(6): p. 2797-801. 81. Yen, B., et al., Molecular basis for ebolavirus VP35 suppression of human dendritic cell maturation. J Virol, 2014. 88(21): p. 12500-10. 82. Alli, R.S. and A. Khar, Interleukin-12 secreted by mature dendritic cells mediates activation of NK cell function. FEBS Lett, 2004. 559(1-3): p. 71-6. 83. Warfield, K.L., et al., Role of natural killer cells in innate protection against lethal ebola virus infection. J Exp Med, 2004. 200(2): p. 169-79. 84. Gupta, M., et al., Monocyte-derived human macrophages and peripheral blood mononuclear cells infected with ebola virus secrete MIP-1alpha and TNF-alpha and inhibit poly-IC-induced IFN-alpha in vitro. Virology, 2001. 284(1): p. 20-5. 85. Stroher, U., et al., Infection and activation of monocytes by Marburg and Ebola viruses. J Virol, 2001. 75(22): p. 11025-33. 86. Hensley, L.E., et al., Proinflammatory response during Ebola virus infection of primate models: possible involvement of the tumor necrosis factor receptor superfamily. Immunol Lett, 2002. 80(3): p. 169-79. 87. Wauquier, N., et al., Human fatal zaire ebola virus infection is associated with an aberrant innate immunity and with massive lymphocyte apoptosis. PLoS Negl Trop Dis, 2010. 4(10). 88. Bradfute, S.B., et al., Lymphocyte death in a mouse model of Ebola virus infection. J Infect Dis, 2007. 196 Suppl 2: p. S296-304. 89. Bradfute, S.B., K.L. Warfield, and S. Bavari, Functional CD8+ T cell responses in lethal Ebola virus infection. J Immunol, 2008. 180(6): p. 4058-66. 90. Maruyama, T., et al., Ebola virus can be effectively neutralized by antibody produced in natural human infection. J Virol, 1999. 73(7): p. 6024-30. 91. Reed, D.S., et al., Depletion of peripheral blood T lymphocytes and NK cells during the course of ebola hemorrhagic Fever in cynomolgus macaques. Viral Immunol, 2004. 17(3): p. 390-400. 92. Yang, Y. and J. Loscalzo, Regulation of tissue factor expression in human microvascular endothelial cells by nitric oxide. Circulation, 2000. 101(18): p. 2144-8. 93. Sanchez, A., et al., Analysis of human peripheral blood samples from fatal and nonfatal cases of Ebola (Sudan) hemorrhagic fever: cellular responses, virus load, and nitric oxide levels. J Virol, 2004. 78(19): p. 10370-7. 94. Kim, P.K., et al., Regulation of caspases by nitric oxide. Ann N Y Acad Sci, 2002. 962: p. 42-52. 95. Bogdan, C., Nitric oxide and the immune response. Nat Immunol, 2001. 2(10): p. 907-16.
  • 30. 30 96. Kroemer, G., The proto-oncogene Bcl-2 and its role in regulating apoptosis. Nat Med, 1997. 3(6): p. 614-20. 97. Lavrik, I., A. Golks, and P.H. Krammer, Death receptor signaling. J Cell Sci, 2005. 118(Pt 2): p. 265-7. 98. Volchkov, V.E., V.M. Blinov, and S.V. Netesov, The envelope glycoprotein of Ebola virus contains an immunosuppressive-like domain similar to oncogenic retroviruses. FEBS Lett, 1992. 305(3): p. 181-4. 99. Sanchez, A., et al., The virion glycoproteins of Ebola viruses are encoded in two reading frames and are expressed through transcriptional editing. Proc Natl Acad Sci U S A, 1996. 93(8): p. 3602-7. 100. Wolf, K., et al., The Ebola virus soluble glycoprotein (sGP) does not affect lymphocyte apoptosis and adhesion to activated endothelium. J Infect Dis, 2011. 204 Suppl 3: p. S947-52. 101. Bray, M. and T.W. Geisbert, Ebola virus: the role of macrophages and dendritic cells in the pathogenesis of Ebola hemorrhagic fever. Int J Biochem Cell Biol, 2005. 37(8): p. 1560-6. 102. Ansari, A.A., Clinical features and pathobiology of Ebolavirus infection. J Autoimmun, 2014. 55: p. 1-9. 103. Wilson, J.A., et al., Epitopes involved in antibody-mediated protection from Ebola virus. Science, 2000. 287(5458): p. 1664-6. 104. Ou, W., et al., Induction of ebolavirus cross-species immunity using retrovirus-like particles bearing the Ebola virus glycoprotein lacking the mucin-like domain. Virol J, 2012. 9: p. 32. 105. Francica, J.R., et al., Steric shielding of surface epitopes and impaired immune recognition induced by the ebola virus glycoprotein. PLoS Pathog, 2010. 6(9): p. e1001098. 106. Marzi, A., et al., Antibodies are necessary for rVSV/ZEBOV-GP-mediated protection against lethal Ebola virus challenge in nonhuman primates. Proc Natl Acad Sci U S A, 2013. 110(5): p. 1893-8. 107. Wong, G., et al., Immune parameters correlate with protection against ebola virus infection in rodents and nonhuman primates. Sci Transl Med, 2012. 4(158): p. 158ra146. 108. Okumura, A., et al., Interaction between Ebola virus glycoprotein and host toll-like receptor 4 leads to induction of proinflammatory cytokines and SOCS1. J Virol, 2010. 84(1): p. 27-33. 109. Martinez, O., C. Valmas, and C.F. Basler, Ebola virus-like particle-induced activation of NF- kappaB and Erk signaling in human dendritic cells requires the glycoprotein mucin domain. Virology, 2007. 364(2): p. 342-54. 110. Druar, C., et al., Analysis of the expressed heavy chain variable-region genes of Macaca fascicularis and isolation of monoclonal antibodies specific for the Ebola virus' soluble glycoprotein. Immunogenetics, 2005. 57(10): p. 730-8. 111. Maruyama, T., et al., Recombinant human monoclonal antibodies to Ebola virus. J Infect Dis, 1999. 179 Suppl 1: p. S235-9. 112. Ito, H., et al., Ebola virus glycoprotein: proteolytic processing, acylation, cell tropism, and detection of neutralizing antibodies. J Virol, 2001. 75(3): p. 1576-80. 113. Yang, Z., et al., Distinct cellular interactions of secreted and transmembrane Ebola virus glycoproteins. Science, 1998. 279(5353): p. 1034-7. 114. Dolnik, O., et al., Ectodomain shedding of the glycoprotein GP of Ebola virus. EMBO J, 2004. 23(10): p. 2175-84. 115. Copelan, E.A., et al., The mechanism of retrovirus suppression of human T cell proliferation in vitro. J Immunol, 1983. 131(4): p. 2017-20. 116. Orosz, C.G., et al., Retrovirus-mediated immunosuppression. I. FeLV-UV and specific FeLV proteins alter T lymphocyte behavior by inducing hyporesponsiveness to lymphokines. J Immunol, 1985. 134(5): p. 3396-403. 117. Cianciolo, G.J., et al., Macrophage accumulation in mice is inhibited by low molecular weight products from murine leukemia viruses. J Immunol, 1980. 124(6): p. 2900-5.
  • 31. 31 118. Yaddanapudi, K., et al., Implication of a retrovirus-like glycoprotein peptide in the immunopathogenesis of Ebola and Marburg viruses. FASEB J, 2006. 20(14): p. 2519-30. 119. Basler, C.F., et al., The Ebola virus VP35 protein functions as a type I IFN antagonist. Proc Natl Acad Sci U S A, 2000. 97(22): p. 12289-94. 120. Cardenas, W.B., et al., Ebola virus VP35 protein binds double-stranded RNA and inhibits alpha/beta interferon production induced by RIG-I signaling. J Virol, 2006. 80(11): p. 5168-78. 121. Prins, K.C., W.B. Cardenas, and C.F. Basler, Ebola virus protein VP35 impairs the function of interferon regulatory factor-activating kinases IKKepsilon and TBK-1. J Virol, 2009. 83(7): p. 3069-77. 122. Basler, C.F., et al., The Ebola virus VP35 protein inhibits activation of interferon regulatory factor 3. J Virol, 2003. 77(14): p. 7945-56. 123. Chang, T.H., et al., Ebola Zaire virus blocks type I interferon production by exploiting the host SUMO modification machinery. PLoS Pathog, 2009. 5(6): p. e1000493. 124. Hay, R.T., SUMO: a history of modification. Mol Cell, 2005. 18(1): p. 1-12. 125. Meulmeester, E. and F. Melchior, Cell biology: SUMO. Nature, 2008. 452(7188): p. 709-11. 126. Feng, Z., et al., The VP35 protein of Ebola virus inhibits the antiviral effect mediated by double-stranded RNA-dependent protein kinase PKR. J Virol, 2007. 81(1): p. 182-92. 127. Reid, S.P., et al., Ebola virus VP24 binds karyopherin alpha1 and blocks STAT1 nuclear accumulation. J Virol, 2006. 80(11): p. 5156-67. 128. Xu, W., et al., Ebola virus VP24 targets a unique NLS binding site on karyopherin alpha 5 to selectively compete with nuclear import of phosphorylated STAT1. Cell Host Microbe, 2014. 16(2): p. 187-200. 129. Reid, S.P., et al., Ebola virus VP24 proteins inhibit the interaction of NPI-1 subfamily karyopherin alpha proteins with activated STAT1. J Virol, 2007. 81(24): p. 13469-77. 130. Platanias, L.C., Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol, 2005. 5(5): p. 375-86. 131. Uddin, S., et al., Activation of the p38 mitogen-activated protein kinase by type I interferons. J Biol Chem, 1999. 274(42): p. 30127-31. 132. Platanias, L.C., The p38 mitogen-activated protein kinase pathway and its role in interferon signaling. Pharmacol Ther, 2003. 98(2): p. 129-42. 133. Halfmann, P., G. Neumann, and Y. Kawaoka, The Ebolavirus VP24 protein blocks phosphorylation of p38 mitogen-activated protein kinase. J Infect Dis, 2011. 204 Suppl 3: p. S953-6. 134. Hoenen, T., A. Groseth, and H. Feldmann, Current ebola vaccines. Expert Opin Biol Ther, 2012. 12(7): p. 859-72. 135. Marzi, A. and H. Feldmann, Ebola virus vaccines: an overview of current approaches. Expert Rev Vaccines, 2014. 13(4): p. 521-31. 136. Mullard, A., Experimental Ebola drugs enter the limelight. Lancet, 2014. 384(9944): p. 649. 137. Ye, L. and C. Yang, Development of vaccines for prevention of Ebola virus infection. Microbes Infect, 2015. 17(2): p. 98-108. 138. Bausch, D.G., et al., Diagnosis and clinical virology of Lassa fever as evaluated by enzyme- linked immunosorbent assay, indirect fluorescent-antibody test, and virus isolation. J Clin Microbiol, 2000. 38(7): p. 2670-7. 139. Leroy, E.M., et al., Diagnosis of Ebola haemorrhagic fever by RT-PCR in an epidemic setting. J Med Virol, 2000. 60(4): p. 463-7. 140. Biel, S.S. and H.R. Gelderblom, Diagnostic electron microscopy is still a timely and rewarding method. J Clin Virol, 1999. 13(1-2): p. 105-19. 141. WHO, WHO Emergency Use Assessment and Listing for Ebola Virus Disease IVDs, Product: ReEBOV™ Antigen Rapid Test Kit. Febuary 2015, WHO.
  • 32. 32 142. Jahrling, P.B., et al., Evaluation of immune globulin and recombinant interferon-alpha2b for treatment of experimental Ebola virus infections. J Infect Dis, 1999. 179 Suppl 1: p. S224-34. 143. Smith, L.M., et al., Interferon-beta therapy prolongs survival in rhesus macaque models of Ebola and Marburg hemorrhagic fever. J Infect Dis, 2013. 208(2): p. 310-8. 144. Geisbert, T.W., et al., Treatment of Ebola virus infection with a recombinant inhibitor of factor VIIa/tissue factor: a study in rhesus monkeys. Lancet, 2003. 362(9400): p. 1953-8. 145. WHO, Categorization and prioritization of drugs for consideration for testing or use in patients infected with Ebola. 2015, World Health Organization: http://who.int/medicines/ebola-treatment/cat_prioritization_drugs_testing/en/. 146. Parren, P.W., et al., Pre- and postexposure prophylaxis of Ebola virus infection in an animal model by passive transfer of a neutralizing human antibody. J Virol, 2002. 76(12): p. 6408-12. 147. Oswald, W.B., et al., Neutralizing antibody fails to impact the course of Ebola virus infection in monkeys. PLoS Pathog, 2007. 3(1): p. e9. 148. Marzi, A., et al., Protective efficacy of neutralizing monoclonal antibodies in a nonhuman primate model of Ebola hemorrhagic fever. PLoS One, 2012. 7(4): p. e36192. 149. Qiu, X., et al., Successful treatment of ebola virus-infected cynomolgus macaques with monoclonal antibodies. Sci Transl Med, 2012. 4(138): p. 138ra81. 150. Olinger, G.G., Jr., et al., Delayed treatment of Ebola virus infection with plant-derived monoclonal antibodies provides protection in rhesus macaques. Proc Natl Acad Sci U S A, 2012. 109(44): p. 18030-5. 151. Qiu, X., et al., Reversion of advanced Ebola virus disease in nonhuman primates with ZMapp. Nature, 2014. 514(7520): p. 47-53. 152. Jiang, S., et al., HIV-1 inhibition by a peptide. Nature, 1993. 365(6442): p. 113. 153. Miller, E.H., et al., Inhibition of Ebola virus entry by a C-peptide targeted to endosomes. J Biol Chem, 2011. 286(18): p. 15854-61. 154. Higgins, C.D., et al., C-peptide inhibitors of Ebola virus glycoprotein-mediated cell entry: effects of conjugation to cholesterol and side chain-side chain crosslinking. Bioorg Med Chem Lett, 2013. 23(19): p. 5356-60. 155. Shah, P.P., et al., A small-molecule oxocarbazate inhibitor of human cathepsin L blocks severe acute respiratory syndrome and ebola pseudotype virus infection into human embryonic kidney 293T cells. Mol Pharmacol, 2010. 78(2): p. 319-24. 156. Cote, M., et al., Small molecule inhibitors reveal Niemann-Pick C1 is essential for Ebola virus infection. Nature, 2011. 477(7364): p. 344-8. 157. Canonico, P.G., et al., In-vivo activity of antivirals against exotic RNA viral infections. J Antimicrob Chemother, 1984. 14 Suppl A: p. 27-41. 158. Huggins, J.W., Prospects for treatment of viral hemorrhagic fevers with ribavirin, a broad- spectrum antiviral drug. Rev Infect Dis, 1989. 11 Suppl 4: p. S750-61. 159. Smither, S.J., et al., Post-exposure efficacy of oral T-705 (Favipiravir) against inhalational Ebola virus infection in a mouse model. Antiviral Res, 2014. 104: p. 153-5. 160. Chimerix, I. Brincidofovir for Ebola. Chimerix, Inc. 2014 [cited 22.11.14. 161. Warren, T.K., et al., Protection against filovirus diseases by a novel broad-spectrum nucleoside analogue BCX4430. Nature, 2014. 508(7496): p. 402-5. 162. Geisbert, T.W., et al., Postexposure protection of guinea pigs against a lethal ebola virus challenge is conferred by RNA interference. J Infect Dis, 2006. 193(12): p. 1650-7. 163. Warren, T.K., et al., Advanced antisense therapies for postexposure protection against lethal filovirus infections. Nat Med, 2010. 16(9): p. 991-4. 164. Grant-Klein, R.J., L.A. Altamura, and C.S. Schmaljohn, Progress in recombinant DNA-derived vaccines for Lassa virus and filoviruses. Virus Res, 2011. 162(1-2): p. 148-61. 165. Pushko, P., et al., Individual and bivalent vaccines based on alphavirus replicons protect guinea pigs against infection with Lassa and Ebola viruses. J Virol, 2001. 75(23): p. 11677-85.
  • 33. 33 166. Sullivan, N.J., et al., Accelerated vaccination for Ebola virus haemorrhagic fever in non-human primates. Nature, 2003. 424(6949): p. 681-4. 167. Bukreyev, A., et al., Successful topical respiratory tract immunization of primates against Ebola virus. J Virol, 2007. 81(12): p. 6379-88. 168. Garbutt, M., et al., Properties of replication-competent vesicular stomatitis virus vectors expressing glycoproteins of filoviruses and arenaviruses. J Virol, 2004. 78(10): p. 5458-65. 169. Feldmann, H., et al., Effective post-exposure treatment of Ebola infection. PLoS Pathog, 2007. 3(1): p. e2. 170. Prins, K.C., et al., Mutations abrogating VP35 interaction with double-stranded RNA render Ebola virus avirulent in guinea pigs. J Virol, 2010. 84(6): p. 3004-15. 171. Warfield, K.L., et al., Ebola virus-like particle-based vaccine protects nonhuman primates against lethal Ebola virus challenge. J Infect Dis, 2007. 196 Suppl 2: p. S430-7. 172. WHO. Case of Marburg Haemorrhagic Fever imported into the Netherlands from Uganda. 2008. 173. CDC, C.f.D.C.a.P. Imported Case of Marburg Hemorrhagic Fever --- Colorado, 2008 2009. 174. Science-Metrix. L’épidémie du virus Ebola et les publications scientifiques – La communauté scientifique réagit rapidement! 2014.
  • 34. 34 Supplementary information: Figure S1 : Worldwide geographic distribution of filovirus hemorrhagic fever cases, 1967–2014. From Martines et all (2015) [50]
  • 35. 35 Figure S2 : Ebola fatal cases and scientific publications on Ebola, 1976- 2014 [174] Figure S3 : Ebola fatal cases and scientific publications on Ebola, March 2014 - October 2014 [174]