You are on page 1of 9

Journal of Clinical Virology 64 (2015) 144152

Contents lists available at ScienceDirect

Journal of Clinical Virology


journal homepage: www.elsevier.com/locate/jcv

Chikungunya, a paradigm of neglected tropical disease that emerged


to be a new health global risk
Virginie Rougeron a,b , I-Ching Sam c , Mlanie Caron a , Dieudonn Nkoghe a , Eric Leroy a,b ,
Pierre Roques d,e,
a

Centre International de Recherches Mdicales de Franceville, Franceville, Gabon


Unit Mixte de Recherche Maladies Infectieuses et Vecteurs: Ecologie, Gntique, Evolution et Contrle (IRD 224 CNRS 5290 UM1-UM2), Institut de
Recherche pour le Dveloppement, Montpellier, France
c
Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
d
CEA, Institute of Emerging Diseases and Innovative Therapies, Division of Immuno-Virology, Fontenay-aux-Roses, France
e
Universit Paris-Sud 11, UMR E1, Orsay, France
b

a r t i c l e

i n f o

Article history:
Received 22 April 2014
Accepted 25 August 2014
Keyword:
Chikungunya

a b s t r a c t
Chikungunya virus (CHIKV) is an alphavirus of the Togaviridae family that causes chronic and incapacitating arthralgia in human populations. Since its discovery in 1952, CHIKV was responsible for sporadic
and infrequent outbreaks. However, since 2005, global Chikungunya outbreaks have occurred, inducing
some fatalities and associated with severe and chronic morbidity. Chikungunya is thus considered as
an important re-emerging public health problem in both tropical and temperate countries, where the
distribution of the Aedes mosquito vectors continues to expand. This review highlights the most recent
advances in our knowledge and understanding of the epidemiology, biology, treatment and vaccination
strategies of CHIKV.
2014 Elsevier B.V. All rights reserved.

1. Introduction
Chikungunya virus (CHIKV) is an alphavirus from the Togaviridae family, which is transmitted by Aedes mosquitoes, and causes
epidemic arthritis or arthralgia together with fever and rash. It is
part of the Semliki Forest virus antigenic complex that also contains ONyong Nyong, Mayaro and Ross River viruses [1]. CHIKV
is a positive-sense, single-stranded RNA virus of about 11.8 kb
(Fig. 1). There are three main genotypes, West African, Central/East
African (C/EA), and Asian, the names reecting the initial geographic restriction of each type. Since CHIKV was rst isolated in
Tanzania in 1952 [2], the disease was mainly conned to localized
outbreaks in Asia and Africa, sometimes with hundreds of thousands of cases [3]. In 2005, a C/EA strain of CHIKV, which likely

Corresponding author at: Service dImmuno-Virologie/UMR E1, Paris XI, Institut


des Maladies Emergentes et Thrapeutiques Innovantes, CEA, B.P.6, 92 265 Fontenay
aux Roses, France. Tel.: +33 01 46 54 91 67; fax: +33 01 46 54 77 26.
E-mail address: pierre.roques@cea.fr (P. Roques).
http://dx.doi.org/10.1016/j.jcv.2014.08.032
1386-6532/ 2014 Elsevier B.V. All rights reserved.

originated in coastal Kenya, spread to islands of the Indian Ocean


and India [4]. Since then, CHIKV spread worldwide to cause large
outbreaks in Southeast and East Asia, as well as many imported
cases in travelers returning to non-endemic regions in Europe and
North America.
This global epidemic affecting millions was unprecedented in
its scale and was likely driven by several factors. These include the
increased volume of travelers, the widening geographic distribution of the mosquito vectors and the adaptation of the epidemic
strain to Aedes albopictus [5]. The susceptibility of mosquitoes in
non-endemic regions such as Australia [6] and North America [7],
and the occurrence of autochthonous outbreaks in Italy [8] and
France [9], have shown that CHIKV can no longer be considered
as a problem of tropical countries. A recent work has also pointed
that CHIKV may be present in severe forms, particularly neurological, which may be fatal [10]. It is also increasingly recognized that
there is a signicant burden of long-term morbidity due to chronic
arthralgia [11]. Consequently, from being a relatively neglected and
understudied pathogen prior to 2004, CHIKV has in recent years
been the focus of intense study.

V. Rougeron et al. / Journal of Clinical Virology 64 (2015) 144152

Fig. 1. Genomic structure of CHIKV and expected function of the produced proteins.
CHIKV has a typical alphavirus genomic organization: 5 -nsP1-nsP2-nsP3-nsP4junction region-C-E3-E2-6K-E1-poly (A)-3 with two open reading frames (ORFs).
The 5 end of the genome has a 7-methylguanosine cap and there is a polyadenylation signal at the 3 end. The 5 ORF is translated from genomic RNA and encodes
four non-structural proteins (nsP1, 2, 3 and 4) encoding for the synthesis of RNA
capping, helicase protease, RNA and RNA dependent RNA polymerase [121]. The 3
ORF is translated from a subgenomic 26S RNA and encodes a polyprotein that is
processed as the capsid protein (C), two surface envelope glycoproteins (E1 and E2),
a small peptide designated E3, and two proteins with ion-channel activity, 6K and
the transframe protein [53,122].

2. Advances in eco-epidemiology (focus: outbreaks and


vector/virus relationship)

145

luteocephalus and Ae. neoafricanus [17]. Other mosquitoes, like Culex


annulirostris and Mansonia africana, have also been incriminated
in CHIKV transmission [14,17,19], and in vitro infection failed to
demonstrate CHIKV infection of Anopheles [21].
In Asia, previous outbreaks occurred mostly in urban areas
and virus maintenance is associated with continuous introduction of the virus in new areas and new nave human populations
[13,2224] (Fig. 2). As in Africa, the major mosquito species
involved was Ae. aegypti. However Ae. albopictus was found during
the recent worldwide outbreaks to be the major vector in several
Asian countries [25,26]. Of concern, the distribution of Ae. albopictus has expanded from South East Asia to Madagascar, the Indian
Ocean islands and Africa, as well as South Europe and USA [27].
In Asian regions, humans have long been considered as the only
reservoirs. However, the recent isolation of CHIKV from monkeys
in Malaysia suggests the potential existence of a sylvatic zoonotic
transmission cycle in Asia [28].

2.1.1. Interactions between reservoirs and vectors

2.1.2. Chikungunya outbreak history

Chikungunya is an arthropod-borne virus transmitted mainly by


Aedes species mosquitoes. Only the female are infective since they
need blood meals for egg formation. Vertical transmission within
mosquitoes is not demonstrated and thus there is no involvement
of the vector in viral persistence in a specic area. However, a recent
study found virus sequences in male mosquitoes, which indirectly
supported trans-ovarian transmission; in addition, males are able
to transmit the virus to females during mating [12]. CHIKV outbreaks often coincide with rainy periods during which increased
mosquitoes densities are observed [13].
In Africa, from where the virus originated, CHIKV transmission
cycle is sylvatic and enzoonotic (Fig. 2). Human infections are due
to occasional spillovers and happen mostly in rural zones, while
urban disease is thought to be due to human migration. Humans
are the principal reservoirs during epidemic periods. Transmission
is mainly ensured by the highly anthropophilic mosquito vectors,
Ae. aegypti and Ae. albopictus [14,15]. During inter-epidemic periods, the transmission cycle appears to be maintained by animal
reservoirs, in particular monkeys. Other reservoirs may include buffalos, rodents, and birds [16] (Fig. 2). Outbreaks occur in monkeys
and animals develop viremia but no pronounced physical manifestations have been observed in the wild [14,17,18]. The principal
sylvatic vectors are Ae. furcifer-taylori [19], Ae. africanus [20], Ae.

Chikungunya was rst identied during an outbreak in


19521953 in East Africa, along the border between Tanzania and
Mozambique (ex-Tanganyika) [2,29]. The name Chikungunya is
derived from a word in the Makonde language that means that
which bends up.
Between 1960 and 1990, Chikungunya outbreaks happened sporadically in Africa and Asia (Fig. 3). In 2004, a major epidemic of the
C/EA strain began in East Africa, specically in Lamu Island and
then in Mombasa, Kenya, before spreading to the Indian Ocean
islands, Western Africa, India, and Asia. Imported CHIKV cases
have been reported in numerous non-endemic countries, including Japan, USA, and Europe. Chikungunya is currently considered
as a real threat in these European and American countries, which
are colonized by Aedes species mosquitoes [30]. The Italian epidemic in 2007 was the rst indigenous outbreak of CHIKV outside
the inter-tropical zone [8]. The recent worldwide outbreaks and
specically the last one in December 2013 in Caribbean Islands, the
Americas, and in Micronesia highlight the important role of international travels in disease and the horizontal exchange of virus and
human populations, which means that the historical segregation
of Asian and African strains together with their ecological specicities are no longer true. The emergence of Chikungunya in mainland
North America is now a reality.
3. Advances in basic virology

Fig. 2. Biological life cycle of Chikungunya virus with interconnection between rural
and urban cycles. In Africa, the CHIKV transmission cycle is considered as sylvatic
and enzoonotic, and human infections are due to occasional spillovers and happen
mostly in rural zones. Urban CHIKV introduction is thought to be due to human
migrations. In Asia, outbreaks occur mostly in urban areas, and virus maintenance
is associated with continuous introduction of the virus into new areas and new
nave human populations. In relation to type of cycle (urban vs. rural), the mosquito
vectors involved are different.

As all alphaviruses (Togaviridae), the chikungunya virion has an


icosahedral capsid enclosed by a lipid envelope and a diameter of
6070 nm. As an enveloped virus, CHIKV is sensitive to desiccation,
temperatures >60 C and detergent.
Beside their role in the viral replication complex, non-structural
proteins are involved in cell control and avoidance of innate
response. Alphaviruses are highly sensitive to the antiviral activity of type I interferons (IFN) [31] and several IFN-stimulated
genes (ISG) including ISG15, ISG20, P56, ZAP, viperin, and 2 ,5 oligoadenylate synthetases (OAS) exert antiviral activity against
them [32,33]. However, alphaviruses have also developed strategies to escape these blocks, and are able to inhibit cellular mRNA
translation. Indeed, their genomic-mRNA contains stem loop structures that allow initiation of translation without requirement of
the cellular translation initiation factor component eIF2a. At the
late stages of the alphavirus life cycle (6 h post-infection), shutoff
of mRNA translation is followed by induction of apoptosis and cell
death [34]. Recent studies showed that balance between autophagy
and apoptosis might play a major role in the capacity of the virus

146

V. Rougeron et al. / Journal of Clinical Virology 64 (2015) 144152

Fig. 3. Geographical distribution of reported outbreaks of chikungunya virus infection in African and Asian regions. Each country presenting CHIKV outbreaks is colored
differently in relation to the outbreaks dates: in green the old outbreaks countries (corresponding to the countries presenting outbreaks only before 2004), in blue countries
touched by outbreaks before and after 2004 and in red the new outbreaks countries (corresponding to the countries where outbreaks occurred only since or after 2004).

to spread during the early stages of the disease [35]. Viral protein recruitment of cell components is prominent, as highlighted
in the selection of ubiquinated capsid by the autophagy receptor p62 promoting cell survival, and the binding of human NDP52
(but not mouse) to nsP2, which promotes viral replication [36].
This may explain the exquisite differences observed in the nonhuman primate and mouse models despite a general involvement
of macrophages in viral persistence [31,3739].
In contrast to new-world alphaviruses, CHIKV RNA packaging
signal was found in the nsP2 sequence, not in nsP1, and is recognized by chikungunya capsid [40]. Moreover, a mutation in nsP2
protease (P718S) showed a role in inhibition of JAK-STAT signaling following IFN type 1 cell activation [41], probably by inhibiting
nsP2 nuclear localization [42]. Another in vitro study, comparing
CHIKV and Sindbis, showed that for CHIKV nsP4, but not Sindbis, the RNA dependent RNA polymerase might specically inhibit
phosphorylation of cellular eIF2a and thus modulate the unfolded
protein response to control the host cells fate [43]. The nsP3 protein
seems to be the most exible protein of the viral genome, as it is the
only place where protein markers can be stably inserted without
modifying CHIKV viral replication [44]. Evolution however seems
to maintain a role in intracellular structure which may vary largely
between alphaviruses and support the need to explore the CHIKV
nsp3 [45]. Specically, CHIKV nsP3 blocks stress granule assembly
by recruitment of G3BP into cytoplasmic foci, and amphiphysin2, which is involved in efcient viral RNA synthesis [46,47]. Some
of these functions are shared within the SFV alphavirus subgroup
[45,47]. Furthermore, nsP3 may play a role in host cell specicity, as
introducing the Onyong nyong nsP3 gene within a CHIKV backbone
allowed a vector shift to Anopheles [21].
In addition to nsP3, motifs in the viral envelope are involved in
the capacity to bind and infect various host cells. Concerning the
vector shift, from Ae. aegypti to Ae. albopictus, so important in the
current outbreaks, the E1-E226V mutation is major, [4851] but
epistatic mutations in E2 (E2-D60G and E2-I211T) may modulate

this effect and even explain the absence of adaptation of the Asian
virus strain to Ae. albopictus [49]. Surprisingly other E1E2 mutant
viruses obtained by truncation of the trans-membrane part of E2
(TMB position AA 374 to 383) remained infectious in mosquito cells
but are attenuated in the vertebrate susceptible host, showing a
complex relationship between infection and pathogenic capacity
[52]. A recent study identied a transframe protein related to the
structural protein 6K which plays a role in particle release in addition to E3 [53,54]. Recently, it was demonstrated that mutants E2
(E2-E166K) in CHIKV viruses, probably found in virus quasi-species,
are able to counteract IFN-induced genes like OAS-3 and ZAP and
in addition trigger a quasi-pure apoptosis phenotype in vitro ([32];
P. Desprs unpublished). Of note, because the packaging of viruses
not only involves the structural proteins but also some of the nonstructural ones, compensatory mutations were observed in nsP
genes as recently described for VEEV nsP2 [42,55].
4. Advances in immuno-virology: mechanisms of
pathogenesis
Following the bite of an infected mosquito, CHIKV is injected
into blood capillaries and dermis. During this intradermal stage,
CHIKV infects the most common cells of connective tissues, epithelial cells and dermal broblasts (Fig. 4). These cells have been shown
to be susceptible to infection and allow viral production [56,57].
Thus viruses produced by both host broblasts and mosquitoes
enter the blood vessels. During the acute phase of infection, CHIKV
antigens can be detected in vivo in blood monocytes [58]. However, it seems that monocytes become a signicant target for CHIKV
only when high levels of virus are present in the circulation. Even
if these cells might be infected, they did not produce substantial
levels of new viruses [58]. This step corresponds to the acute stage
of the CHIKV infection, which can last up to 12 days [59]. At this
stage, viral load can be extremely high, at >108 copies/mL [60]
(Figs. 4 and 5). Because of their extensive distribution among the

V. Rougeron et al. / Journal of Clinical Virology 64 (2015) 144152

147

Fig. 4. Chikungunya infection pathogenesis model. Following the bite of an infected mosquito, CHIKV is injected into blood capillaries and dermis. During this intradermal
stage, CHIKV infects human epithelial cells and dermal broblasts. Blood monocytes and macrophages then become infected with CHIKV and viral replication happens
(>108 copies/m). Monocytes are then responsible for viral dissemination and systemic infection. The principal secondary infection sites are muscles and joints, endothelial
cells of the liver and the brain and macrophages and stroma cells of the spleen and lymph nodes. Immune responses are also induced, with high production of IFN- and
liberation of several cytokines and chemokines.

different organs and tissues and their migration capacity after


activation through blood and lymphatic systems, monocytes are
thought to be responsible for viral dissemination and systemic
infection (Fig. 4). The principal secondary infection sites are muscles and joints, where endothelial cells, broblasts and satellite cells
are infected. Endothelial cells of the liver and the brain are also
infected, leading to possible hepatitis and encephalopathy. Finally,
macrophages and stroma cells of the spleen and the lymph nodes
are infected too, and, together with joint tissues, may be sites of
virus persistence [38,39].
During viral infection, innate immunity is rst activated to suppress virus propagation, replication and dissemination. There is a
large and rapid production of several pro-inammatory cytokines,
chemokines and growth factors [61] (Figs. 4 and 5). IFN-/ play a
key early role in controlling the severity of viral infections including
alphavirus infections [6265]. IFN-/ sub-types trigger signaling
pathways including the expression of hundreds of interferonstimulated genes, resulting in the synthesis of multiple antiviral
proteins and the establishment of an overall antiviral state. IFN has been shown to be produced readily in vitro and after CHIKV
infection in humans [58,66], particularly by CHIKV-infected broblasts [67]. IFN- levels have been shown to be normal or marginally
increased [38,66]. During the acute phase, in parallel with this

IFN-type 1 liberation, a strong inammatory response is observed,


at rst with production of IL-6, MCP-1, IP-10 and RANTES. These
chemokines are involved in the recruitment of leucocytes to the
infection sites and in the deployment of efcient antiviral defenses
as well as high inammatory response although levels vary with
the cohort studied [6870]. There is also induction of a strong cellular immune response, during which IFN-, IL-4, IL-7 and other
cytokines involved in adaptive immunity are released [61]. The proinammatory cytokine IL-12 has also been detected rapidly after
infection with CHIKV [58,71]. Both IFN- and IL-12 activate natural
killer (NK) cells, a major component of the innate immune defense.
Specic subsets of NK cells are able to sense CHIKV from the beginning of the infection and may thus contribute to viral clearance
[61,72]. The B-cell promoting cytokines IL-4 and IL-10 are upregulated during the rst days after symptoms onset, and may initiate
CHIKV-specic IgG production [61,73] (Fig. 5). This is followed by
the activation of adaptive responses through CD8+ T cells. Most of
these observations were conrmed in non-human primate model
of the infection [39]. Finally, a switch of CD8+ T cells to CD4+ T cells
activation and the production of IL-1R and IL-2RA [61] characterize the late acute stages. CD4+ T cells are the major mediator of
inammation in the joint swelling and tissue damage in an acute
mouse model [74].

148

V. Rougeron et al. / Journal of Clinical Virology 64 (2015) 144152

Fig. 5. Chikungunya virus infection evolution and clinical diagnosis. After transmission of CHIKV through an infected mosquito bite, a period of incubation begins for
24 days on average. Then, an abrupt clinical onset is observed which lasts between
7 and 10 days. During this time, viral load may be extremely high, exceeding 108 viral
copies/ml of blood, and is followed by activation of innate immunity leading to high
levels of interferon (IFN). Anti-CHIKV antibodies can be detected in patients shortly
after symptoms onset, usually from 4 to 6 days for IgM, and remain detectable for
several weeks to several months. IgG are detectable only a few days later (715
days), and persist for life.

The role of cytokine in chronic stage is far less understood [38],


but the role of infected osteoblasts and joint macrophages in the
persistence of the disease might be associated with a vicious loop
of IL6/RankL production [68,75,76].
5. Advances in clinical virology
5.1.1. Clinical presentation
Following an incubation period of 24 days (range 112 days)
after an infective mosquito bite, Chikungunya infection is characterized by the classical and self-limiting symptoms of an abrupt
onset of fever (38 C), macular rash, and arthralgia [77,78].
However, the recent outbreaks enabled close scrutiny of large
numbers of patients, leading to reports of atypical and severe manifestations, sometimes fatal (reviewed by Rajapakse [79]). These
include myocarditis [10], extensive bullous skin lesions [80], and
encephalopathy [10]. Neonatal encephalopathy was also described
following vertical transmission at birth [81]. Chronic arthralgia,
which may be incapacitating, causes long-term morbidity and have
high economic impact. This may be due to defective clearance and
subsequent persistence of CHIKV antigen in macrophages in joints,
stimulating ongoing inammation [68]. Reported rates of chronic
arthralgia vary between countries, from 0 to 4.1% at 3 months in
Gabon [80] and Malaysia [82], to 60% at 3 years in La Runion [83],
which may reect differences in underlying population genetics,
healthcare practices, or CHIKV strain virulence. The most commonly reported risk factor for chronic arthralgia is older age [83,84].
In endemic countries, acute Chikungunya disease may be difcult to distinguish clinically from other febrile illnesses such as
dengue and malaria [80,84], although certain specic features such
as acute red ears have been described [85]. Therefore, laboratory
diagnosis remains important.
5.1.2. Laboratory diagnosis
There have been notable advances in CHIKV diagnostics over
the last few years. The gold standard methods for diagnosis are
virus isolation and nucleic acid amplication tests, although these

are not widely available in most endemic countries. Both methods are most useful in the rst 7 days of acute symptoms, when
viremia is present before IgM becomes detectable (Fig. 5). Numerous conventional and real-time RT-PCR assays have been described
[60,86]. Peak viremia levels are seen at days 13 of symptoms and
RNA from non-viable virus can still be detected in the presence of
antibodies as late as day 12 [60]. Apart from increased sensitivity
compared to conventional PCR, real-time PCR also allows viral load
quantication, which warrants further study as a potential indicator of prognosis. At present, conicting data exist as to whether
higher viral loads are associated with increased [38] or reduced risk
of persistent arthralgia [87], or if there is no link [83]. An alternative
for diagnosis during the early stages of infection is antigen detection, of which there are two reports, [88], [89] although further
evaluation of performance is required.
CHIKV patients often do not present early in infection; in Sri
Lanka and Malaysia, patients presented at a median of 45 days [90]
(Sam, unpublished). The serological detection of IgM is thus important. However, the limited data available on use of commercial kits
in a clinical setting are disappointing. A commercial immunouorescence assay had a sensitivity of 76% at day 5 and 100% at
day 7 [91], but most diagnostic laboratories in endemic areas are
unlikely to have a uorescence microscope. Other ELISA and rapid
immunochromatographic tests (ICT) performed poorly, with sensitivity rates of 022% before day 7, and 1783% after day 7, when
retrospective diagnosis is of less clinical value [90,92], [93]. Furthermore, performance of the tests is affected by the match between
the antigen used in the assay and the circulating CHIKV strain, presumably leading to differences in IgM binding. In Singapore, an ICT
using recombinant E1-226A had a sensitivity of 77% for samples collected at days 57 during an outbreak of CHIKV bearing E1-226A;
during a later outbreak of CHIKV with E1-226V, sensitivity fell to
0% [91]. The use of CHIKV antigens of C/EA origin in ICTs may also
explain the poor sensitivities of 2053% in Indonesia, where the
Asian genotype of CHIKV was circulating [92]. Currently available
CHIKV IgM kits therefore are either unsuited, or cannot be solely
relied upon for diagnosis of acute infection.
There remains a need for simple, affordable, sensitive and specic diagnostic tests for CHIKV in developing countries, for the
different periods over which a patient may present, preferably at
the point of care. For early presentations of <7 days, an RNA/antigen
detection assay would be appropriate. Loop-mediated isothermal
amplication is a possible option for resource-limited settings,
as it can be performed in a single tube at a single temperature
in a water-bath, with greater sensitivity than conventional PCR
[94]. For presentations after >5 days, an IgM assay is necessary
[86]. Recently, patients antibody responses to CHIKV proteins have
been characterized, showing that linear epitopes at the N terminus
of E2 glycoprotein induce early antibodies that persist, although
binding may be affected by a K252Q change which can occur
naturally [95]. Further work is needed to identify and validate wellconserved antigens that may be reliably used for serological assays.
All new assays will then require subsequent testing with wellcharacterized samples in different settings with genetically diverse
circulating strains.

6. Advances in prophylaxis and therapy


Development of Chikungunya vaccine began as early as 1967
but after the relative failure of the US-Army vaccine in 1980,
interest and efforts waned until the recent re-emergence of the
disease. [68]. Currently, no licensed vaccine against Chikungunya is
commercially available, but numerous candidates are under study
(reviewed by Thiberville [96]). The virus-like particle (VLP) candidates are of most interest [9799], one of which is currently

V. Rougeron et al. / Journal of Clinical Virology 64 (2015) 144152

149

undergoing safety evaluation [100]. Only few vaccines have been


tested in non-human primates or phase 1/2 studies in humans
[101], but most seem efcient in a mouse model as showed by our
European group [102,103], and future deployment depends more
on economic measures rather than technical barriers [52,98].
Passive immunotherapy was historically used against viral disease. Some studies showed that extracted IgG against CHIKV
might protect against infection [97,104]. This protection may
also be achieved with human recombinant monoclonal antibodies [105,106]. Although these proofs of concept were obtained in
mice, such strategies might be very valuable to treat acute severe
diseases such as infection in babies [104].
As CHIKV disease was previously relatively rare, specic drugs
acting against CHIKV are not available. Molecules, known to have
non-specic antiviral action like chloroquine and arbidol, showed
some efciency in vitro [107,108], but no clinical efcacy [109]. In
addition, using the non-human primate model that we have developed in CEA, we demonstrated that chloroquine administration
during the acute phase or before infection seems to enhance viral
replication and may have adverse effects on the number of chronic
cases [110].
Screening of crude extracts of tropical plants revealed molecules
(trigocherrins, trigowiin) with good selectivity index in vitro against
CHIKV. However, their in vivo toxicity, tumorigenicity, and mechanism of action still remain to be dened [111,112]. Using a similar
method to screen a library of 502 highly puried natural product
compounds, harringtonine, a cephalotaxine alkaloid, was found as
acting as a CHIKV inhibitor [113]. In a high throughput screen using
uorescent CHIKV replicons of 356 clinically approved drugs, 5
potential inhibitors of CHIKV were also discovered [114].
Previously the demonstration of the role of cellular furinprotease in the processing of envelope proteins of Chikungunya
was reinforced by specic inhibition of these proteins [115]. Nevertheless, usage in clinical assay does not seem relevant as targeting
the viral protease (nsP2- or capsid-associated protease) should
be more specic and hopefully will induce fewer side effects.
Thus by combining structure-based specic computing, an in silico screen of molecules able to interfere with the nsP2 protease site
allowed selection of 4 putative candidates [116], for future in cellulo
experiments. Hence, a screen of 3040 molecules against the nsP2
mediated shut-off using a luciferase-based reporter gene identied only one natural product able to inhibit specically CHIKV
replication in cellulo [117]. Recently, targeting the natural cell
response against alphavirus [67] by inducing RIG-1 using an optimized 5 triphosphorylated RNA in vitro inhibited viral replication
[118].
Finally, few experimental treatments targeting host response
or host components involved during Chikungunya cell infection
have been tested in animal models. However, in a mouse model of
Ross River virus-induced arthritis, the demonstrated effects of an
inhibitor of macrophage migration inhibitory factor action [119]
deserve to be evaluated in pre-clinical assay, especially because
of the analogy of the CHIKV pathophysiology in both mouse and
macaque models [120].
To date, none of these treatments have been used in humans
either in the acute or chronic phases of disease, due to lack of effort
to develop relevant animal models of the chronic disease despite
our rst promising results [39].

reservoirs should be investigated to assess their involvement in


transmission in endemic areas and the associated risk in newly
exposed countries.
The most impressive advances have been made in basic virology. Up to 2005, most of the data for CHIKV were deduced from
studies of other alphaviruses. The role of epistatic mutations in
adaptation to host cells was an important discovery that might
help to understand specic pathogenic effects of the virus in mammalian or mosquito cells. This may aid in future viral surveillance
for potential epidemic threats. Moreover, this will assist in designing attenuated viruses for vaccination, currently the least expensive
mode of vaccine design. Another important advance was the use of
Chikungunya human clinical strains with a focus on low passaged
viruses. Finally, understanding the cellular cycle in vivo, the role
of the various cellular targets and the interplay between viral and
cellular proteins in the clinical evolution of CHIKV patients will provide tools for both diagnosis and treatment. In particular, there is
a real need for simple and affordable diagnostic tests for endemic
developing countries.
The understanding of the pathogenesis of acute CHIKV improved
these last years, and several cytokines and chemokines have been
explored and suggested as playing a key role in the interactions
between CHIKV and the host immune system. However, exploring
how the innate immune system, in particular the role of NK cells,
affects the outcome of CHIKV infection should be considered in the
future. Interestingly, the nature and the severity of CHIKV infection and of cytokine proles vary between human populations with
the percentage of chronic disease. In this context, it would also be
of great interest to focus future research on the study of human
genetic resistance.
Chikungunya fever is currently symptomatically treated with
antipyretics and non-steroidal anti-inammatory drugs. There will
be a continued search for new antiviral candidates with a clearly
dened mechanism of viral inhibition in cell-based systems and
signicant activity in animal models. These will include therapy
based on specic immunoglobulins or molecules that modify the
inammatory response associated with CHIKV infection. Moreover,
studies on the pathogenesis of Chikungunya chronic rheumatic
manifestations and the effects of disease-modifying anti-rheumatic
drugs should be explored, as the burden of chronic disease has been
shown to be unexpectedly high. Standardization of chronic patient
follow-ups and animal models are needed for this purpose.

7. 10 years research roadmap

Acknowledgments

The recent global outbreaks of CHIKV have driven great


advances in understanding of the disease. Interestingly, CHIKV or its
specic antibodies have been detected in several mammals other
than primates. The poorly understood role of these intermediate

The authors are grateful to Dr. Philippe Desprs for helpful comments and for providing unpublished data. We are also thankful to
Caroline Petitdemange from CIRMF for help in editing the text and
gures.

Funding
PR and ICS received funding from the European Unions Seventh Framework Program ICRES (Grant Agreement No. 261202).
ICS was funded by University Malaya (UMRG grant RG526-13HTM
and PPP Grant PG030-2012B) and the Ministry of Higher Education,
Malaysia (FRGS Grant FP036-2013A).
Competing interests
The authors declare they have no competing interest.
Ethical approval
Not required.

150

V. Rougeron et al. / Journal of Clinical Virology 64 (2015) 144152

References
[1] Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. Chikungunya
virus and prospects for a vaccine. Expert Rev Vaccines 2005;11:1087101.
[2] Robinson MC. An epidemic of virus disease in Southern Province, Tanganyika Territory, in 195253. I. Clinical features. Trans R Soc Trop Med Hyg
1955;49:2832.
[3] De Ranitz CM, Myers RM, Varkey MJ, Isaac ZH, Carey DE. Clinical impressions
of chikungunya in Vellore gained from study of adult patients. Indian J Med
Res 1965;53:75663.
[4] Kariuki Njenga M, Nderitu L, Ledermann JP, Ndirangu A, Logue CH, Kelly CH,
et al. Tracking epidemic Chikungunya virus into the Indian Ocean from East
Africa. J Gen Virol 2008;89:275460.
[5] Tsetsarkin KA, Vanlandingham DL, McGee CE, Higgs S. A single mutation
in chikungunya virus affects vector specicity and epidemic potential. PLoS
Pathog 2007;3:e201.
[6] van den Hurk AF, Hall-Mendelin S, Pyke AT, Smith GA, Mackenzie JS. Vector
competence of Australian mosquitoes for chikungunya virus. Vector Borne
Zoonotic Dis 2010;10:48995.
[7] Reiskind MH, Pesko K, Westbrook CJ, Mores CN. Susceptibility of Florida
mosquitoes to infection with chikungunya virus. Am J Trop Med Hyg
2008;78:4225.
[8] Rezza G, Nicoletti L, Angelini R, Romi R, Finarelli AC, Panning M, et al. Infection
with chikungunya virus in Italy: an outbreak in a temperate region. Lancet
2007;370:18406.
[9] Grandadam M, Caro V, Plumet S, Thiberge JM, Souares Y, Failloux AB,
et al. Chikungunya virus, southeastern France. Emerg Infect Dis 2011;17:
9103.
[10] Lemant J, Boisson V, Winer A, Thibault L, Andre H, Tixier F, et al. Serious acute
chikungunya virus infection requiring intensive care during the Reunion
Island outbreak in 20052006. Crit Care Med 2008;36:253641.
[11] Sissoko D, Malvy D, Ezzedine K, Renault P, Moscetti F, Ledrans M, et al.
Post-epidemic Chikungunya disease on Reunion Island: course of rheumatic
manifestations and associated factors over a 15-month period. PLoS Negl Trop
Dis 2009;3:e389.
[12] Mavale M, Parashar D, Sudeep A, Gokhale M, Ghodke Y, Geevarghese G, et al.
Venereal transmission of chikungunya virus by Aedes aegypti mosquitoes
(Diptera: Culicidae). Am J Trop Med Hyg 2010;83:12424.
[13] Powers AM, Logue CH. Changing patterns of chikungunya virus: re-emergence
of a zoonotic arbovirus. J Gen Virol 2007;88:236377.
[14] Jupp PG, McIntosh BM, Dos Santos I, DeMoor P. Laboratory vector studies on
six mosquito and one tick species with chikungunya virus. Trans R Soc Trop
Med Hyg 1981;75:159.
[15] Rao TR. Vectors of dengue and chikungunya viruses: a brief review. Indian J
Med Res 1964;52:71926.
[16] Kading RC, Borland EM, Craneld M, Powers AM. Prevalence of antibodies to alphaviruses and aviviruses in free-ranging game animals and
nonhuman primates in the greater congo basin. J Wildl Dis 2013;49:
58799.
[17] Diallo M, Thonnon J, Traore-Lamizana M, Fontenille D. Vectors of Chikungunya
virus in Senegal: current data and transmission cycles. Am J Trop Med Hyg
1999;60:2816.
[18] Binn LN, Harrison VR, Randall R. Patterns of viremia and antibody observed in
rhesus monkeys inoculated with chikungunya and other serologically related
group A arboviruses. Am J Trop Med Hyg 1967;16:7825.
[19] Jupp PG, McIntosh BM. Aedes furcifer and other mosquitoes as vectors of
chikungunya virus at Mica, northeastern Transvaal, South Africa. J Am Mosq
Control Assoc 1990;6:41520.
[20] Powers AM, Brault AC, Tesh RB, Weaver SC. Re-emergence of Chikungunya
and Onyong-nyong viruses: evidence for distinct geographical lineages and
distant evolutionary relationships. J Gen Virol 2000;81:4719.
[21] Saxton-Shaw KD, Ledermann JP, Borland EM, Stovall JL, Mossel EC, Singh AJ,
et al. Onyong nyong virus molecular determinants of unique vector specicity
reside in non-structural protein 3. PLoS Negl Trop Dis 2013;7:e1931.
[22] Chevillon C, Briant L, Renaud F, Devaux C. The Chikungunya threat:
an ecological and evolutionary perspective. Trends Microbiol 2008;16:
808.
[23] Laras K, Sukri NC, Larasati RP, Bangs MJ, Kosim R. Djauzi, et al. Tracking the
re-emergence of epidemic chikungunya virus in Indonesia. Trans R Soc Trop
Med Hyg 2005;99:12841.
[24] Lam SK, Chua KB, Hooi PS, Rahimah MA, Kumari S, Tharmaratnam M, et al.
Chikungunya infection an emerging disease in Malaysia. Southeast Asian J
Trop Med Public Health 2001;32:44751.
[25] Das B, Sahu A, Das M, Patra A, Dwibedi B, Kar SK, et al. Molecular investigations
of chikungunya virus during outbreaks in Orissa, Eastern India in 2010. Infect
Genet Evol 2012;12:1094101.
[26] Thavara U, Tawatsin A, Pengsakul T, Bhakdeenuan P, Chanama S, Anantapreecha S, et al. Outbreak of chikungunya fever in Thailand and virus
detection in eld population of vector mosquitoes, Aedes aegypti (L.) and
Aedes albopictus Skuse (Diptera: Culicidae). Southeast Asian J Trop Med Public
Health 2009;40:95162.
[27] Gratz NG. Critical review of the vector status of Aedes albopictus. Med Vet
Entomol 2004;18:21527.
[28] Apandi Y, Lau SK, Izmawati N, Amal NM, Faudzi Y, Mansor W, et al. Identication of Chikungunya virus strains circulating in Kelantan, Malaysia in 2009.
Southeast Asian J Trop Med Public Health 2010;41:137480.

[29] Ross RW. The Newala epidemic. III. The virus: isolation, pathogenic properties
and relationship to the epidemic. J Hyg (Lond) 1956;54:17791.
[30] Boutin JP, Simon F, Oliver M, Grandadam M, Queyriaux B, Tolou H. [Marseilles, the Indian Ocean and Chikungunya virus]. Bull Acad Natl Med
2007;191:7857.
[31] Couderc T, Chretien F, Schilte C, Disson O, Brigitte M, Guivel-Benhassine F,
et al. A mouse model for Chikungunya: young age and inefcient type-I interferon signaling are risk factors for severe disease. PLoS Pathog 2008;4:e29.
[32] Henrik Gad H, Paulous S, Belarbi E, Diancourt L, Drosten C, Kummerer BM,
et al. The E2-E166K substitution restores Chikungunya virus growth in OAS3
expressing cells by acting on viral entry. Virology 2012;434:2737.
[33] Teng TS, Foo SS, Simarmata D, Lum FM, Teo TH, Lulla A, et al. Viperin controls chikungunya-virus replication and pathology in humans and mice. J Clin
Invest 2012;122:444760.
[34] Krejbich-Trotot P, Gay B, Li-Pat-Yuen G, Hoarau JJ, Jaffar-Bandjee MC, Briant
L, et al. Chikungunya triggers an autophagic process which promotes viral
replication. Virol J 2011;8:432.
[35] Joubert PE, Werneke SW, de la Calle C, Guivel-Benhassine F, Giodini A, Peduto
L, et al. Chikungunya virus-induced autophagy delays caspase-dependent cell
death. J Exp Med 2012;209:102947.
[36] Judith D, Mostowy S, Bourai M, Gangneux N, Lelek M, Lucas-Hourani M, et al.
Species-specic impact of the autophagy machinery on Chikungunya virus
infection. EMBO Rep 2013;14:53444.
[37] Gardner J, Anraku I, Le TT, Larcher T, Major L, Roques P, et al. Chikungunya
virus arthritis in adult wild-type mice. J Virol 2010;84:802132.
[38] Hoarau JJ, Jaffar Bandjee MC, Krejbich Trotot P, Das T, Li-Pat-Yuen G, Dassa B,
et al. Persistent chronic inammation and infection by Chikungunya arthritogenic alphavirus in spite of a robust host immune response. J Immunol
2010;184:591427.
[39] Labadie K, Larcher T, Joubert C, Mannioui A, Delache B, Brochard P, et al.
Chikungunya disease in nonhuman primates involves long-term viral persistence in macrophages. J Clin Invest 2010;120:894906.
[40] Kim DY, Atasheva S, Foy NJ, Wang E, Frolova EI, Weaver S, et al. Design of
chimeric alphaviruses with a programmed, attenuated, cell type-restricted
phenotype. J Virol 2011;85:436376.
[41] Fros JJ, Liu WJ, Prow NA, Geertsema C, Ligtenberg M, Vanlandingham DL,
et al. Chikungunya virus nonstructural protein 2 inhibits type I/II interferonstimulated JAK-STAT signaling. J Virol 2010;84:1087787.
[42] Fros JJ, van der Maten E, Vlak JM, Pijlman GP. The C-terminal domain of chikungunya virus nsP2 independently governs viral RNA replication, cytopathicity
and inhibition of interferon signaling. J Virol 2013;87:10394400.
[43] Rathore AP, Ng ML, Vasudevan SG. Differential unfolded protein response
during Chikungunya and Sindbis virus infection: CHIKV nsP4 suppresses
eIF2alpha phosphorylation. Virol J 2013;10:36.
[44] Kummerer BM, Grywna K, Glasker S, Wieseler J, Drosten C. Construction of
an infectious Chikungunya virus cDNA clone and stable insertion of mCherry
reporter genes at two different sites. J Gen Virol 2012;93:19915.
[45] Foy NJ, Akhrymuk M, Akhrymuk I, Atasheva S, Bopda-Waffo A, Frolov I,
et al. Hypervariable domains of nsP3 proteins of New World and Old World
alphaviruses mediate formation of distinct, virus-specic protein complexes.
J Virol 2013;87:19972010.
[46] Fros JJ, Domeradzka NE, Baggen J, Geertsema C, Flipse J, Vlak JM, et al. Chikungunya virus nsP3 blocks stress granule assembly by recruitment of G3BP into
cytoplasmic foci. J Virol 2012;86:108739.
[47] Neuvonen M, Kazlauskas A, Martikainen M, Hinkkanen A, Ahola T, Saksela K.
SH3 domain-mediated recruitment of host cell amphiphysins by alphavirus
nsP3 promotes viral RNA replication. PLoS Pathog 2011;7:e1002383.
[48] Schuffenecker I, Iteman I, Michault A, Murri S, Frangeul L, Vaney MC, et al.
Genome microevolution of chikungunya viruses causing the Indian Ocean
outbreak. PLoS Med 2006;3:e263.
[49] Tsetsarkin KA, Chen R, Leal G, Forrester N, Higgs S, Huang J, et al. Chikungunya virus emergence is constrained in Asia by lineage-specic adaptive
landscapes. Proc Natl Acad Sci U S A 2011;108:78727.
[50] Tsetsarkin KA, McGee CE, Volk SM, Vanlandingham DL, Weaver SC, Higgs S.
Epistatic roles of E2 glycoprotein mutations in adaption of chikungunya virus
to Aedes albopictus and Ae. aegypti mosquitoes. PLoS ONE 2009;4:e6835.
[51] Vazeille M, Moutailler S, Coudrier D, Rousseaux C, Khun H, Huerre M, et al.
Two Chikungunya isolates from the outbreak of La Reunion (Indian Ocean)
exhibit different patterns of infection in the mosquito, Aedes albopictus. PLoS
ONE 2007;2:e1168.
[52] Piper A, Ribeiro M, Smith KM, Briggs CM, Huitt E, Nanda K, et al. Chikungunya
virus host range E2 transmembrane deletion mutants induce protective
immunity against challenge in C57BL/6J mice. J Virol 2013;87:674857.
[53] Snyder JE, Kulcsar KA, Schultz KL, Riley CP, Neary JT, Marr S, et al. Functional
characterization of the alphavirus TF protein. J Virol 2013;87:851123.
[54] Uchime O, Fields W, Kielian M. The role of E3 in pH protection during
alphavirus assembly and exit. J Virol 2013;87:1025562.
[55] Kim DY, Firth AE, Atasheva S, Frolova EI, Frolov I. Conservation of a packaging signal and the viral genome RNA packaging mechanism in alphavirus
evolution. J Virol 2011;85:802236.
[56] Thon-Hon VG, Denizot M, Li-Pat-Yuen G, Giry C, Jaffar-Bandjee MC, Gasque
P. Deciphering the differential response of two human broblast cell lines
following Chikungunya virus infection. Virol J 2012;9:213.
[57] Sourisseau M, Schilte C, Casartelli N, Trouillet C, Guivel-Benhassine F, Rudnicka D, et al. Characterization of reemerging chikungunya virus. PLoS Pathog
2007;3:e89.

V. Rougeron et al. / Journal of Clinical Virology 64 (2015) 144152


[58] Her Z, Malleret B, Chan M, Ong EK, Wong SC, Kwek DJ, et al. Active infection
of human blood monocytes by Chikungunya virus triggers an innate immune
response. J Immunol 2010;184:590313.
[59] Das T, Jaffar-Bandjee MC, Hoarau JJ, Krejbich Trotot P, Denizot M, LeePat-Yuen G, et al. Chikungunya fever: CNS infection and pathologies of a
re-emerging arbovirus. Prog Neurobiol 2010;91:1219.
[60] Laurent P, Le Roux K, Grivard P, Bertil G, Naze F, Picard M, et al. Development of
a sensitive real-time reverse transcriptase PCR assay with an internal control
to detect and quantify chikungunya virus. Clin Chem 2007;53:140814.
[61] Wauquier N, Becquart P, Nkoghe D, Padilla C, Ndjoyi-Mbiguino A, Leroy
EM. The acute phase of Chikungunya virus infection in humans is associated with strong innate immunity and T CD8 cell activation. J Infect Dis
2011;204:11523.
[62] Sen A, Rothenberg ME, Mukherjee G, Feng N, Kalisky T, Nair N, et al. Innate
immune response to homologous rotavirus infection in the small intestinal villous epithelium at single-cell resolution. Proc Natl Acad Sci U S A
2007;109:2066772.
[63] Ryman KD, Meier KC, Gardner CL, Adegboyega PA, Klimstra WB. Nonpathogenic Sindbis virus causes hemorrhagic fever in the absence of
alpha/beta and gamma interferons. Virology 2007;368:27385.
[64] Briolant S, Garin D, Scaramozzino N, Jouan A, Crance JM. In vitro inhibition of
Chikungunya and Semliki Forest viruses replication by antiviral compounds:
synergistic effect of interferon-alpha and ribavirin combination. Antiviral Res
2004;61:1117.
[65] Gardner CL, Burke CW, Higgs ST, Klimstra WB, Ryman KD. Interferonalpha/beta deciency greatly exacerbates arthritogenic disease in
mice infected with wild-type chikungunya virus but not with the cell
culture-adapted live-attenuated 181/25 vaccine candidate. Virology
2012;425:10312.
[66] Ng LF, Chow A, Sun YJ, Kwek DJ, Lim PL, Dimatatac F, et al. IL-1beta, IL-6, and
RANTES as biomarkers of Chikungunya severity. PLoS ONE 2009;4:e4261.
[67] Schilte C, Couderc T, Chretien F, Sourisseau M, Gangneux N, GuivelBenhassine F, et al. Type I IFN controls chikungunya virus via its action on
nonhematopoietic cells. J Exp Med 2010;207:42942.
[68] Dupuis-Maguiraga L, Noret M, Brun S, Le Grand R, Gras G, Roques P. Chikungunya disease: infection-associated markers from the acute to the chronic
phase of arbovirus-induced arthralgia. PLoS Negl Trop Dis 2012;6:e1446.
[69] Chirathaworn C, Poovorawan Y, Lertmaharit S, Wuttirattanakowit N. Cytokine
levels in patients with chikungunya virus infection. Asian Pac J Trop Med
2013;6:6314.
[70] Reddy V, Mani RS, Desai A, Ravi V. Correlation of plasma viral loads and presence of chikungunya IgM antibodies to cytokine/chemokine levels during
acute chikungunya virus infection. J Med Virol 2014;86:1393401.
[71] Roques P, Gras G. Chikungunya fever: focus on peripheral markers of pathogenesis. J Infect Dis 2011;203:1413.
[72] Petitdemange C, Becquart P, Wauquier N, Beziat V, Debre P, Leroy EM, et al.
Unconventional repertoire prole is imprinted during acute chikungunya
infection for natural killer cells polarization toward cytotoxicity. PLoS Pathog
2011;7:e1002268.
[73] Kam YW, Lum FM, Teo TH, Lee WW, Simarmata D, Harjanto S, et al. Early
neutralizing IgG response to Chikungunya virus in infected patients targets a dominant linear epitope on the E2 glycoprotein. EMBO Mol Med
2012;4:33043.
[74] Teo TH, Lum FM, Claser C, Lulla V, Lulla A, Merits A, et al. A pathogenic
role for CD4+ T cells during Chikungunya virus infection in mice. J Immunol
2013;190:25969.
[75] Noret M, Herrero L, Rulli N, Rolph M, Smith PN, Li RW, et al. Interleukin
6, RANKL, and osteoprotegerin expression by chikungunya virus-infected
human osteoblasts. J Infect Dis 2012;206:4557, 579.
[76] Phuklia W, Kasisith J, Modhiran N, Rodpai E, Thannagith M, Thongsakulprasert T, et al. Osteoclastogenesis induced by CHIKV-infected broblast-like
synoviocytes: a possible interplay between synoviocytes and monocytes/macrophages in CHIKV-induced arthralgia/arthritis. Virus Res
2013;177:17988.
[77] Taubitz W, Cramer JP, Kapaun A, Pfeffer M, Drosten C, Dobler G, et al. Chikungunya fever in travelers: clinical presentation and course. Clin Infect Dis
2007;45:e14.
[78] Nimmannitya S, Mansuwan P. Comparative clinical and laboratory ndings
in conrmed dengue and chikungunya infections. Bull World Health Organ
1966;35:423.
[79] Rajapakse S, Rodrigo C, Rajapakse A. Atypical manifestations of chikungunya
infection. Trans R Soc Trop Med Hyg 2010;104:8996.
[80] Nkoghe D, Kassa RF, Caron M, Grard G, Mombo I, Bikie B, et al. Clinical forms
of chikungunya in Gabon, 2010. PLoS Negl Trop Dis 2012;6:e1517.
[81] Gerardin P, Barau G, Michault A, Bintner M, Randrianaivo H, Choker G, et al.
Multidisciplinary prospective study of mother-to-child chikungunya virus
infections on the island of La Reunion. PLoS Med 2008;5:e60.
[82] Ayu SM, Lai LR, Chan YF, Hatim A, Hairi NN, Ayob A, et al. Seroprevalence
survey of Chikungunya virus in Bagan Panchor, Malaysia. Am J Trop Med Hyg
2010;83:12458. United States.
[83] Schilte C, Staikovsky F, Couderc T, Madec Y, Carpentier F, Kassab S, et al.
Chikungunya virus-associated long-term arthralgia: a 36-month prospective
longitudinal study. PLoS Negl Trop Dis 2013;7:e2137.
[84] Mohd Zim MA, Sam IC, Omar SF, Chan YF, AbuBakar S, Kamarulzaman A.
Chikungunya infection in Malaysia: comparison with dengue infection in
adults and predictors of persistent arthralgia. J Clin Virol 2013;56:1415.

151

[85] Javelle E, Tiong TH, Leparc-Goffart I, Savini H, Simon F. Inammation of the


external ear in acute chikungunya infection: experience from the outbreak in
Johor Bahru, Malaysia, 2008. J Clin Virol 2014;59:2703.
[86] Reddy V, Ravi V, Desai A, Parida M, Powers AM, Johnson BW. Utility of IgM
ELISA, TaqMan real-time PCR, reverse transcription PCR, and RT-LAMP assay
for the diagnosis of Chikungunya fever. J Med Virol 2012;84:17718.
[87] Kam YW, Simarmata D, Chow A, Her Z, Teng TS, Ong EK, et al. Early
appearance of neutralizing immunoglobulin G3 antibodies is associated with
chikungunya virus clearance and long-term clinical protection. J Infect Dis
2012;205:114754.
[88] Kashyap RS, Morey SH, Chandak NH, Purohit HJ, Taori GM, Daginawala HF.
Detection of viral antigen, IgM and IgG antibodies in cerebrospinal uid of
Chikungunya patients with neurological complications. Cerebrospinal Fluid
Res 2010;7:12.
[89] Shukla J, Khan M, Tiwari M, Sannarangaiah S, Sharma S, Rao PV, et al. Development and evaluation of antigen capture ELISA for early clinical diagnosis
of chikungunya. Diagn Microbiol Infect Dis 2009;65:1429.
[90] Blacksell SD, Tanganuchitcharnchai A, Jarman RG, Gibbons RV, Paris DH, Bailey MS, et al. Poor diagnostic accuracy of commercial antibody-based assays
for the diagnosis of acute Chikungunya infection. Clin Vaccine Immunol
2011;18:17735.
[91] Yap G, Pok KY, Lai YL, Hapuarachchi HC, Chow A, Leo YS, et al. Evaluation of
Chikungunya diagnostic assays: differences in sensitivity of serology assays
in two independent outbreaks. PLoS Negl Trop Dis 2010;4:e753.
[92] Kosasih H, Widjaja S, Surya E, Hadiwijaya SH, Butarbutar DP, Jaya UA, et al.
Evaluation of two IgM rapid immunochromatographic tests during circulation
of Asian lineage Chikungunya virus. Southeast Asian J Trop Med Public Health
2012;43:5561.
[93] Rianthavorn P, Wuttirattanakowit N, Prianantathavorn K, Limpaphayom N,
Theamboonlers A, Poovorawan Y. Evaluation of a rapid assay for detection
of IgM antibodies to chikungunya. Southeast Asian J Trop Med Public Health
2010;41:926.
[94] Parida MM, Santhosh SR, Dash PK, Tripathi NK, Lakshmi V, Mamidi N,
et al. Rapid and real-time detection of Chikungunya virus by reverse transcription loop-mediated isothermal amplication assay. J Clin Microbiol
2007;45:3517.
[95] Kam YW, Lee WW, Simarmata D, Harjanto S, Teng TS, Tolou H, et al. Longitudinal analysis of the human antibody response to chikungunya virus
infection: implications for serodiagnosis and vaccine development. J Virol
2012;86:1300515.
[96] Thiberville SD, Moyen N, Dupuis-Maguiraga L, Antoine N, Gould EA, Roques P,
et al. Chikungunya fever: epidemiology, clinical syndrome, pathogenesis and
therapy. Antiviral Res 2013;99:34570.
[97] Akahata W, Yang ZY, Andersen H, Sun S, Holdaway HA, Kong WP, et al. A
virus-like particle vaccine for epidemic Chikungunya virus protects nonhuman primates against infection. Nat Med 2010;16:3348.
[98] Metz SW, Gardner J, Geertsema C, Le TT, Goh L, Vlak JM, et al. Effective chikungunya virus-like particle vaccine produced in insect cells. PLoS Negl Trop Dis
2013;7:e2124.
[99] Brandler S, Rufe C, Combredet C, Brault JB, Najburg V, Prevost MC, et al. A
recombinant measles vaccine expressing chikungunya virus-like particles is
strongly immunogenic and protects mice from lethal challenge with chikungunya virus. Vaccine 2013;31:371825.
[100] Paneez Khoury PI. Chikungunya Virus Vaccine Trial in Healthy Adults. VRC
311: A Phase 1 Open Label, Dose-Escalation Clinical Trial to Evaluate the Safety
and Immunogenicity of a Virus-Like Particle (VLP) Chikungunya Vaccine,
VRC-CHKVLP059-00-VP. In: Healthy Adults. National Institute of Allergy and
Infectious Diseases (NIAID); 2011. ClinicalTrialsgov identier: NCT01489358.
[101] Roy CJ, Adams AP, Wang E, Plante K, Gorchakov R, Seymour RL, et al.
Chikungunya vaccine candidate is highly attenuated and protects nonhuman
primates against telemetrically-monitored disease following a single dose. J
Infect Dis 2014;209:18919.
[102] Garcia-Arriaza J, Cepeda V, Hallengard D, Sorzano CO, Kummerer BM, Liljestrom P, et al. A novel poxvirus-based vaccine (MVA-CHIKV) is highly
immunogenic and protects mice against chikungunya infection. J Virol
2014;88:352747.
[103] Hallengard D, Kakoulidou M, Lulla A, Kummerer BM, Johansson DX, Mutso
M, et al. Novel attenuated Chikungunya vaccine candidates elicit protective
immunity in C57BL/6 mice. J Virol 2014;88:285866.
[104] Couderc T, Khandoudi N, Grandadam M, Visse C, Gangneux N, Bagot S,
et al. Prophylaxis and therapy for Chikungunya virus infection. J Infect Dis
2009;200:51623.
[105] Fric J, Bertin-Maghit S, Wang CI, Nardin A, Warter L. Use of human monoclonal antibodies to treat Chikungunya virus infection. J Infect Dis 2013;207:
31922.
[106] Pal P, Dowd KA, Brien JD, Edeling MA, Gorlatov S, Johnson S, et al. Development of a highly protective combination monoclonal antibody therapy
against Chikungunya virus. PLoS Pathogens 2013;9:e1003312.
[107] Delogu I, de Lamballerie X. Chikungunya disease and chloroquine treatment.
J Med Virol 2011;83:10589.
[108] Delogu I, Pastorino B, Baronti C, Nougairede A, Bonnet E, de Lamballerie X.
In vitro antiviral activity of arbidol against Chikungunya virus and characteristics of a selected resistant mutant. Antiviral Res 2011;90:99107.
[109] De Lamballerie X, Boisson V, Reynier JC, Enault S, Charrel RN, Flahault A, et al.
On chikungunya acute infection and chloroquine treatment. Vector Borne
Zoonotic Dis 2008;8:8379.

152

V. Rougeron et al. / Journal of Clinical Virology 64 (2015) 144152

[110] Roques P. Experimental infection of macaques and pathophysiology of


Chikungunya infection. York Hotel, Singapore: Forum about Challenges
and insights towards understanding the reemergence of chikungunya;
2009.
[111] Allard PM, Martin MT, Dau ME, Leyssen P, Gueritte F, Litaudon M, et al. the
rst natural chlorinated daphnane diterpene orthoester from Trigonostemon
cherrieri. Org Lett 2012;14:3425.
[112] Bourjot M, Leyssen P, Eydoux C, Guillemot JC, Canard B, Rasoanaivo P, et al.
Flacourtosides A-F, phenolic glycosides isolated from Flacourtia ramontchi. J
Nat Prod 2012;75:7528.
[113] Kaur P, Thiruchelvan M, Lee RC, Chen H, Chen KC, Ng ML, et al. Inhibition of chikungunya virus replication by harringtonine, a novel antiviral
that suppresses viral protein expression. Antimicrob Agents Chemother
2013;57:15567.
[114] Pohjala L, Utt A, Varjak M, Lulla A, Merits A, Ahola T, et al. Inhibitors of
alphavirus entry and replication identied with a stable Chikungunya replicon cell line and virus-based assays. PLoS ONE 2011;6:e28923.
[115] Ozden S, Lucas-Hourani M, Ceccaldi PE, Basak A, Valentine M, Benjannet S,
et al. Inhibition of Chikungunya virus infection in cultured human muscle
cells by furin inhibitors: impairment of the maturation of the E2 surface
glycoprotein. J Biol Chem 2008;283:21899908.
[116] Singh Kh D, Kirubakaran P, Nagarajan S, Sakkiah S, Muthusamy K, Velmurgan D, et al. Homology modeling, molecular dynamics, e-pharmacophore

[117]

[118]

[119]

[120]

[121]
[122]

mapping and docking study of Chikungunya virus nsP2 protease. J Mol Model
2012;18:3951.
Lucas-Hourani M, Lupan A, Despres P, Thoret S, Pamlard O, Dubois J, et al.
A phenotypic assay to identify Chikungunya virus inhibitors targeting the
nonstructural protein nsP2. J Biomol Screen 2013;18:1729.
Olagnier D, Scholte FE, Chiang C, Albulescu IC, Nichols C, He Z, et al.
Inhibition of dengue and chikungunya virus infection by RIG-I-mediated
type I IFN-independent stimulation of the innate antiviral response. J Virol
2014;88:418094.
Herrero LJ, Nelson M, Srikiatkhachorn A, Gu R, Anantapreecha S, FingerleRowson G, et al. Critical role for macrophage migration inhibitory factor (MIF)
in Ross River virus-induced arthritis and myositis. Proc Natl Acad Sci U S A
2011;108:1204853.
Dupuis-Maguiraga L, Larcher T, Vezmar K, Dereuddre-Bosquet N, Babarit C,
Delache B, et al. Arthrogenic alphaviruses: comparison between Chikungunya
and Ross River virus in non-human primates. In: 11th arbovirus research in
Australia and the 10th mosquito control association of Australia symposium
Surfer Paradise, Gold Coast, Queensland, Australia, September 9-14, 2012.
Jose J, Snyder JE, Kuhn RJ. A structural and functional perspective of alphavirus
replication and assembly. Future Microbiol 2009;4:83756.
Voss JE, Vaney MC, Duquerroy S, Vonrhein C, Girard-Blanc C, Crublet E, et al.
Glycoprotein organization of Chikungunya virus particles revealed by X-ray
crystallography. Nature 2010;468:70912.

You might also like