Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Médecine et maladies infectieuses 49 (2019) 574–585

Disponible en ligne sur

ScienceDirect
www.sciencedirect.com

Review

Crimean-Congo hemorrhagic fever: An update


Fièvre hémorragique de Crimée-Congo: mise au point
P. Fillâtre a,∗ , M. Revest b , P. Tattevin b
a
Service de réanimation polyvalente, centre hospitalier St-Brieuc, 10, rue Marcel-Proust, 22000 St-Brieuc, France
b
Service des maladies infectieuses et de réanimation médicale, CHU de Rennes, Rennes, France

a r t i c l e i n f o a b s t r a c t

Article history: Crimean-Congo hemorrhagic fever (CCHF) is a severe form of hemorrhagic fever caused by a virus of the
Received 7 September 2018 genus Nairovirus. The amplifying hosts are various mammal species that remain asymptomatic. Humans
Received in revised form 22 October 2018 are infected by tick bites or contact with animal blood. CCHF has a broad geographic distribution and is
Accepted 3 September 2019
endemic in Africa, Asia (in particular the Middle East) and South East Europe. This area has expanded
Available online 10 October 2019
in recent years with two indigenous cases reported in Spain in 2016 and 2018. The incubation period is
short with the onset of symptoms in generally less than a week. The initial symptoms are common to
Keywords:
other infectious syndromes with fever, headache, myalgia and gastrointestinal symptoms. The hemor-
Crimean-Congo hemorrhagic fever
Viral hemorrhagic fever
rhagic syndrome occurs during a second phase with sometimes major bleeding in and from the mucous
Health professionals membranes and the skin. Strict barrier precautionary measures are required to prevent secondary and
Ribavirin nosocomial spread. CCHF may be documented by PCR detection of the virus genome during the first days
Ticks after the onset of illness, and then by serological testing for IgM antibodies as from the 2nd week after
infection. Patient management is mainly based on supportive care. Despite a few encouraging retrospec-
tive reports, there is no confirmed evidence that supports the use of ribavirin for curative treatment.
Nevertheless, the World Health Organization continues to recommend the use of ribavirin to treat CCHF,
considering the limited medical risk related to short-term treatment. The prescription of ribavirin should
however be encouraged post-exposure for medical professionals, to prevent secondary infection.
© 2019 Elsevier Masson SAS. All rights reserved.

r é s u m é

Mots clés : La fièvre hémorragique de Crimée-Congo (FHCC) est une forme sévère de fièvre hémorragique causée
Fièvre hémorragique de Crimée-Congo par un virus du genre Nairovirus. De nombreux mammifères peuvent être hôtes intermédiaires et restent
Fièvre hémorragique virale asymptomatiques. Les humains sont infectés par morsures de tiques ou par le contact avec du sang
Professionnels de santé animal. La FHCC a une large distribution géographique et est endémique en Afrique, en Asie (en particulier
Ribavirine
au Moyen-Orient) et dans le Sud-Est de l’Europe. Cette zone s’est étendue ces dernières années avec
Tiques
deux cas autochtones signalés en Espagne en 2016 et 2018. La période d’incubation est courte et les
symptômes apparaissent généralement en moins d’une semaine. Les symptômes initiaux sont communs
à d’autres syndromes infectieux tels que fièvre, céphalées, myalgies et troubles gastro-intestinaux. Le
syndrome hémorragique survient au cours d’une seconde phase avec parfois des saignements importants
cutanéomuqueux. Des mesures d’isolement strictes sont nécessaires pour prévenir les cas secondaires
nosocomiaux. La FHCC peut être documentée par détection en PCR du génome au cours des premiers
jours suivant l’apparition de la maladie, puis par sérologique avec la recherche d’anticorps IgM à compter
de la 2e semaine après l’infection. La gestion des patients repose principalement sur des traitements
symptomatiques. Malgré quelques séries rétrospectives encourageantes, il n’existe pas de haut niveau
de preuve en faveur de l’utilisation de la ribavirine pour le traitement curatif. Néanmoins, l’Organisation
mondiale de la santé continue de recommander l’utilisation de la ribavirine dans le traitement de la FHCC,
compte tenu du risque limité sur des traitements de courte durée. La prescription de ribavirine devrait
toutefois être encouragée en post-exposition pour les professionnels de la santé, afin de prévenir les
infections secondaires.
© 2019 Elsevier Masson SAS. Tous droits réservés.

∗ Corresponding author.
E-mail address: pierre.fillatre@ch-stbrieuc.fr (P. Fillâtre).
https://doi.org/10.1016/j.medmal.2019.09.005
0399-077X/© 2019 Elsevier Masson SAS. All rights reserved.
P. Fillâtre et al. / Médecine et maladies infectieuses 49 (2019) 574–585 575

1. Introduction 4. Taxonomy and phylogenetics

Crimean-Congo hemorrhagic fever (CCHF) virus is one of the The CCHF virus belongs to the genus Nairovirus, of the order
most severe viral diseases for humans. Typically, the disease is ini- Bunyaviridae. The Nairovirus genus comprises 34 different viruses
tially characterized by unspecific signs and symptoms before the divided into 7 distinct serological subgroups. Only 3 viruses of this
onset of the hemorrhagic syndrome. A fatality rate of 5–30% is gen- genus can cause disease in humans, CCHF being of greater impor-
erally reported [1]. The present literature review discusses the most tance than Dugbe fever and Nairobi sheep fever [2]. The nairovirus
recently published data on the epidemiology, mode of transmis- genome is structured into 3 segments of single-stranded RNA,
sion, clinical forms and potential treatment options for the disease, named L, M, and S based on their respective sizes, that are packaged
bearing in mind a possible outbreak of CCHF in mainland France. in an internal envelop formed by the NP nucleoprotein. The diame-
ter of the virion is approximately 90–120 nm. The virion consists of
a lipid envelop comprising the GN and GC glycoproteins which play
2. Material and methods a key role in the cell adhesion process prior to endocytosis, hemag-
glutination and inducing a host immune response [1,6]. Because
The PubMed database was searched with the term “Crimean the virus is highly contagious and causes severe disease, it should
Congo hemorrhagic fever” and the following filters: 1/articles in only be handled in high-containment (BSL-4) laboratories.
English and French, and 2/articles published between January 1, Studies on the antigenic structure of the CCHF viral mem-
2000 and September 1, 2018. In all, 971 references were identified. brane could not detect any differences between the Crimean and
The number of articles used for this literature review was further African strains before 1967. Today, the viral RNA (in particular
reduced by selection based on the title and abstract. the S segment) has been sequenced and 8 genetic lineages have
been identified. The first European lineage comprises the viruses
found in East Europe covering a region from the Balkan peninsula,
3. History of CCHF through Turkey and into Russia [2]. A second lineage, named AP92
and found mainly in Greece, forms an independent group and is
Although several cases of illness that can probably be assimi- probably less pathogenic. The third lineage is present in Central
lated to CCHF were described in Tajikistan from the 12th century Asia (Kazakhstan, Tajikistan, Uzbekistan and China), and the fourth
on, the first documented reports of the disease were described in in Pakistan, Madagascar and some of the strains found in Iran. The
1944 when the Soviet armed forces engaged the Germany army fifth lineage consists of the remaining strains found in Iran, as well
in Crimea [2]. Nearly 200 soldiers fell ill with a fever associated as those found in Senegal and Mauritania. The other three lineages
with hemorrhagic syndrome and shock, with a mortality rate of are found only in Africa [1].
10% [1,3]. The disease was suspected to be due to a tick-borne
virus following the injection of a filtered suspension of ticks, at
the same time as antibiotics, to volunteer soldiers and psychiatric 5. Epidemiology
patients [2]. The virus was not identified until 1967, when Chu-
makov, a Soviet scientist, isolated it in a patient from Uzbekistan CCHF has the most widespread geographical distribution of all
[4]. the tick-borne viruses that cause disease in humans, and the second
Independently, another virus causing similar clinical fea- widest geographical distribution of all arboviruses, after Dengue
tures was isolated in 1956 from a child in the Democratic virus [1] (Fig. 1). During the 20th century, the main CCHF outbreaks
Republic of Congo. Only in 1969 were the Congo and Uzbek- occurred in the ex-Soviet Union (e.g. Crimea, Rostov, Astrakhan), in
istan strains described as displaying antigenic similarity. The Bulgaria with 1105 cases reported between 1953 and 1974 [7], and
virus was then renamed Crimean-Congo hemorrhagic fever virus in China where 260 farmers were infected between 1965 and 1994
[2,5]. in the Xinjiang region, with a reported mortality of 80% (probably

Fig. 1. Risk areas for Crimean-Congo hemorrhagic fever (CCHF), adapted from [119].
Zones à risque de fièvre hémorragique Crimée-Congo (FHCC), issue de [119].
576 P. Fillâtre et al. / Médecine et maladies infectieuses 49 (2019) 574–585

Table 1
Cases of Crimean-Congo hemorrhagic fever (CCHF) reported since 2000.
Cas de fièvre hémorragique Crimée-Congo (FHCC) publiés depuis 2000.

Country Screening method Period Reference Description

Europe
Kosovo – 2001 [120] 75 suspected cases, 5 deaths
Albania Serology 2001 [121] 8 cases, including 1 secondary case in a health professional
Bulgaria – 2000–2003 [7] 98 cases reported to the Ministry of Health over 4 years
Serology and PCR 2008 [122] 4 cases, including 1 secondary case in a health professional
PCR 2014 [123] 1 imported case in the United Kingdom from Bulgaria
Greece PCR 2008 [124] 1 death, a farmer from North-East Greece
Spain PCR 2016 [28] 1st indigenous case in Western Europe, and a secondary case in a
[29] health professional
2nd indigenous case in Spain due to a tick bite, Castile and Leon region
Georgia PCR 2009 [125] 1st case reported in the Tbilissi region
Serology 2008–2011 [126] 3 cases detected among 537 patients included for fever > 38 ◦ C and no
Serology and PCR 2014 [127] etiology after > 48 hours
22 cases reported to the Ministry of Health between January and
September 2014
Middle East
Turkey Serology and PCR 2002–2003 [128] 29 documented cases (9 PCR+, 25 IgM+) in the Black Sea region
PCR 2007 [129] 10 imported cases in Istanbul
– 2002–2017 [5] 10,000 cases reported to the Ministry of Health over 15 years.
Reported mortality rate: 4.6%
Iran – 2004 [130] 6 cases in a province in the North of Iran
Serology and PCR 1996–2006 [131] Pediatric series of 34 children and adolescents. Reported mortality
Serology and PCR 2000–2007 [132] rate: 26% (9/34)
397 cases confirmed by the Pasteur Institute in Tehran
Iraq Serology and PCR 1998–2010 [133] 43 confirmed cases over 13 years, mostly in the Ninawa province
Oman Serology 2011 [134] 1st confirmed case in Oman
Dubai PCR 2010 [135] 2 reported cases
Asia
Pakistan Serology and PCR 1997–2002 [136] 83 confirmed cases (9 PCR+, 74 seropositives), Balochistan province
Antigen, serology and 2005 [137] 8 cases in Karachi. Reported mortality rate: 75%
PCR 2005 [138] 1 case, Abbottabad province
PCR 2011 [139] 8 cases, Hazara province
– 2009 [140] 1 case in a 16-year boy in Karachi
PCR 2012 [141] 1 imported case in Glasgow (Scotland) in a patient who stayed in
PCR 2009 [142] Kabul for 3 weeks
PCR 1 imported case in Germany in a US soldier previously stationed in the
south of Afghanistan
Kazakhstan Serology and PCR 2000–2013 [36] 212 confirmed cases over 14 years
India PCR 2011 [143] First series of 5 confirmed cases in India
Africa
Kenya PCR 2002 [144] 1 isolated case
Senegal Serology and PCR 2003 [145] 1 isolated case
PCR 2004 [52] 1 imported case in Rennes (France) from Dakar
Mauritania Serology and PCR 2003 [146] 38 cases, including 15 secondary cases in health professionals
Sudan PCR 2008 [147] 8 confirmed cases including 4 secondary hospital-acquired cases
PCR 2009 [148] Outbreak in 7 patients in a region of South Sudan
Uganda PCR 2015 [149] 1 isolated case

biased because only the most severe cases were recorded) [1]. Most Sudan [12] and reflect an important reservoir on the African con-
of the cases described in the literature since January 1, 2020 are tinent. In Europe, the virus has been found in livestock in Bulgaria,
listed in Table 1. Currently, the main outbreak sites are found in the Hungary, Albania, Kosovo and Greece, although at lower levels, and
Middle East (Turkey and Iran), with over 10,000 cases documented even in two bats in the south of France [13,14].
in Turkey since 2002 [5]. The vector of the disease is ticks of the genus Hyalomma, which
A large number of studies have been carried out to determine are found in Asia and Africa but also in Europe (Fig. 1). They have
the seropositivity rate and delimit the geographical area where the been found on migrating birds in Italy [15], Germany [16], in the
virus is found. High seropositivity rates have been found in rumi- south of France [17] and Spain [18]. The virus was first detected
nants (sheep, cows, goats), the amplifying hosts of CCHF and act in ticks collected on deer in Spain in 2002 [19]. The CCHF virus
as reliable indicators of the presence of the virus in some regions. detected was of African lineage and was phylogenetically related
There is a logical relationship between a high seropositivity rate in to the lineage found in ticks that infest migratory birds in Morocco
animals in a given region and the incidence of the disease in humans [20]. To date, however, only very few ticks in Spain have been found
[8]. Indeed, in a study performed on 1,165 ruminants in Bulgaria and to carry the virus, between 0 and 3,2% [5,18].
Turkey, seropositivity rates of 26% and 57% were reported, respec-
tively, with major divergences from region to region: absence of 6. CCHF virus life cycle
seropositivity in some western regions of Turkey but a seroposi-
tivity rate of up to 87% in regions near the Black Sea [8]. In Africa, 6.1. Ticks: host and vector
although compared with the Middle East less human cases of CCHF
are reported, the seropositivity rates are sometimes high in live- The CCHF virus has been found in about thirty different species
stock with 1.6% in the Democratic Republic of Congo [9], 66% in of ticks (Fig. 2). The most efficient vector belongs to the Ixodes genus,
Mali [10], 67% in Mauritania [11] and 21% in dromedary camels in the Hyalomma marginatum tick, because it can:
P. Fillâtre et al. / Médecine et maladies infectieuses 49 (2019) 574–585 577

Fig. 2. Transmission of Crimean-Congo hemorrhagic fever, adapted from [5,21].


Transmission de la fièvre hémorragique de Crimée-Congo, adapté de [5,21].

• acquire the virus when feeding on an infected host. Inside the tick, hot seasons that enable the ticks to develop from larvae to adults,
the virus replicates within the tick’s intestinal wall and spreads to as well as the favorable arid environment and vegetation found
various tissue types, reaching the highest titers in salivary glands in Mediterranean countries with large populations of both small
and reproductive organs; and large mammals, it is possible that the incidence of CCHF will
• transmit the virus vertically during its various life stages (larva, increase in upcoming years and spread to yet unaffected regions
nymph and adult); [24]. Some authors even suggest that the future climate change
• transmit the virus via its offspring via the transovarial pathway in and spreading distribution area of the Hyalomma ticks, will result
females. A female produces several thousand eggs, so even a low in the emergence of CCHF in western Europe [25–27], with a num-
transovarial transmission rate is likely to maintain a relatively ber of cases far greater that the indigenous cases described in Spain
large population of infected ticks; in 2016 and August 2018 [28,29]. In this setting, the CCHF virus
• and transmit the virus horizontally when several ticks are present was classified as a priority zoonotic infection 10 years ago by Santé
on a same host animal because infected ticks introduce the virus Publique France (French Health Authority) that should be closely
into the host’s bloodstream immediately upon biting. monitored and investigated for efficient preventive and disease
control measures [30].
To fully engorge, female ticks can remain for several weeks on a
host. The likelihood of transmitting the virus to other ticks that feed 7. Transmission to humans
on the same animal is therefore increased [3,6,21]. Some Hyalomma
marginatum ticks carry the virus throughout their various life stages 7.1. Livestock farming
and therefore are true natural CCHF reservoirs. Other tick species,
such as ticks of the Argasid genus, cannot maintain and transmit Human infection usually occurs by a tick bite or by squashing
the virus horizontally and vertically [3,22]. an infected tick with ungloved hands. Despite potential recall bias
and the fact that some tick bites can be missed if the tick remains
6.2. Amplifying hosts only for a short time on the host, tick bites are reported by 60 to
69% of patients [31,32]. An increase in the number of cases seems
Vertebrates are the amplifying hosts of CCHF and develop high to be recorded in the spring and the summer when adult Hyalomma
titers of the virus in less than 2 weeks following infection [21], ticks require to feed to complete their life cycle [23], especially if
however there have been no reports of the virus causing symp- the previous winter was mild causing an increased survival rate of
toms in animals as of yet. Seropositivity rate investigations have infected ticks [33]. The capacity of Hyalomma ticks to transmit the
shown that large herbivores are most frequently infected. Birds CCHF virus to humans also depends on the ecosystem in which they
generally do not develop the virus, with the exception of ostriches develop. In regions where there are large numbers of both small
[3]. However, birds contribute to spreading the disease by carrying mammals such as hares and hedgehogs and large mammals such
CCHF-infected ticks over hundreds of kilometers [23]. Small verte- as cattle and sheep, the virus seems to circulate silently with only
brates such as hares and hedgehogs are important for maintaining a few sporadic “accidental” human cases. In contrast, as seemed to
virus populations because often infested with larval stage ticks that have been the case in Crimea in 1944 when wild hares had pro-
then transmit the virus vertically during their different life stages. liferated on abandoned farms during the German occupation and
Large vertebrates that can be infested with several dozens of ticks at the number of farm animals had dropped significantly, the soldiers
the same time, can also amplify the virus by horizontal transmission and farm workers that repopulated the area indirectly represented
[6,21]. a new source of blood for the adult Hyalomma ticks, resulting in
Due to [1] the very broad geographical distribution of the a high incidence of human CCHF cases [3]. It is possible that the
Hyalomma vector, [2] the fact that a great many species of verte- same kind of circumstances could explain the outbreaks seen in
brates can act as amplifying hosts, and [3] the dry climate and long some regions of Turkey [23].
578 P. Fillâtre et al. / Médecine et maladies infectieuses 49 (2019) 574–585

Humans can also be infected by direct contact with animal blood 8. Preventive measures
or other infected tissue, although muscle acidification following
death of the animal inactivates the virus. In a study describing a 8.1. For travelers
series of 1820 cases registered in Turkey between 2002 and 2007,
patients reported that they had had close contact with animals in The risk of contracting the disease during travel to an endemic
62% of the cases [32]. In Turkey, nearly 90% of the cases reported area, and therefore of importing the disease back to Europe, is con-
occurred in farmers, slaughterhouse workers or butchers [1]. It sidered to be very low. A comprehensive literature review revealed
has also been shown that the seropositivity rate for anti-CCHF that only 21 cases of such imported disease were reported between
IgG antibodies is higher in elderly patients, as well as patients of 1960 and 2016, albeit with 12 fatalities. Two cases were imported
lower social-economic status, with a history of tick bites, regularly into Western Europe from Bulgaria and three cases from Africa
remove ticks from animals, handle livestock, slaughter animals or [51]. Only one of these cases occurred in France, in Rennes at
process meat and globally in contact with animals in endemic areas the end of 2004, and was imported from Senegal, a country for
[23,34–37]. Human cases are predominantly observed in men, but which no outbreaks were reported at that time [52]. Neverthe-
this should be seen in correlation with farming activities that are less, it seems important that people traveling to endemic areas
mostly performed by men in the countries the most at risk, as in be properly informed about vector control measures against tick-
the Middle East. borne diseases (repellents, wearing long clothing), the need to wear
The possible sexual transmission of CCHF should also be consid- gloves to remove ticks, the risks of contamination via contact with
ered, although very few cases have been documented [38]. Several livestock and working with meat.
cases of mother-to-child transmission have been reported, with
severe hemorrhagic syndromes in neonates and subsequent death 8.2. For health professionals
[5,39], although in other cases neonates are not infected which
suggests that the virus is not transmitted vertically in all cases In mainland France, a patient returning from travel to an
[40]. endemic area with fever and thrombocytopenia is considerably
more likely to have contracted malaria or Dengue fever. A failure
to consider a diagnosis of CCHF would result in a risk of a hospi-
7.2. Transmission to health care professionals tal outbreak of the disease if adequate isolation and barrier nursing
techniques are not implemented. Therefore, the French operational
The second category of people most affected by CCHF infec- group for coordinating epidemic and biological risks (Coordination
tion are health care professionals and laboratory workers. A great opérationnelle en réseau du risque épidémique et biologique [COREB]),
many such cases have been described [28,41–46] with a few related together with expert advice from the infectious diseases depart-
deaths. A large retrospective study was performed on 9 sites in ments of reference health facilities, has recently issued guidelines
Turkey between 2002 and 2014 [47]. During that period, health pro- for 1st-line health care providers for the management of patients
fessionals cared for 4869 patients with confirmed CCHF infection. with suspected viral hemorrhagic fever [53]. These recommenda-
There were 51 cases of accidental exposure of health professionals tions are in line with European guidelines on infection control in
over that period with 25 confirmed secondary cases and a mortal- the management of highly pathogenic infectious diseases [54].
ity rate of 16%. In most cases, infection was due to a needlestick The first crucial step for managing patients with CCHF is
injury (62.7%) followed by contact between infected biological flu- to consider its diagnosis, which must result in the immediate
ids and the health professional’s mucous membrane (23.5%). The implementation of specific barrier precautions. Standard barrier
transmission rate following a needlestick injury was estimated precautions seem sufficient for patients in the early stages of the
at 25%. Post-exposure prophylactic protocols with oral ribavirin disease, with a first level of isolation via the use of an alcohol-
were implemented in the event of needlestick injuries with a based hand sanitizer, level 2 face mask, disposable gown and non
certain amount of success. For a total of 32 needlestick injuries, sterile gloves. If the patient presents with body excretions (vom-
no (0) secondary cases were seen in the 19 carers who received iting, profuse diarrhea, significant hemorrhagic syndrome, etc.)
prophylactic treatment, whereas 8 of the 13 carers who did not then protective measures should be augmented with a splash-
receive prophylactic treatment developed the disease. Neverthe- resistant face mask, single-use scrub suits, a full length waterproof
less, although certain experts recommend prophylactic treatment gown/coverall, two pairs of non sterile nitrile gloves and eyeglasses
with ribavirin, no well-documented studies have yet validated its with side shields [53]. The patient should be cared for in a separate
use [48]. isolation room, away from the main patient flow, because cases
The risk of transmission is maximal during the hemorrhagic of transmission in double patient rooms and to visitors have been
phase, i.e. the late stages of the disease, whereas no cases of sec- described [54,55]. If possible, the isolation room should have neg-
ondary infection have been reported during the incubation phase. ative room pressure. Waste should be managed specifically and
The risk of contamination is highest in the event of needlestick incinerated. When the recommended confinement measures are
injuries and an absence of protective measures to avoid expo- properly implemented, the risk of secondary infection in health
sure to blood and other infectious body fluids during care or professionals is probably insignificant [56]. This is supported by
washing of a deceased patient [49]. In Russia, cases have been the absence of seroconversion in any of the health professionals
documented in health professionals who were not in direct con- who cared for the unique case of imported CCHF in France in 2004,
tact with biological fluids, although the study did not describe despite confinement measures being initially limited to avoiding
in detail possible contacts with contaminated materials. These direct contact [52,57,58].
findings raise the question of possible airborne transmission, but
such cases seem only to occur on an exceptional basis [50]. This 8.3. Vaccination
mode of transmission has already been described for viral hemor-
rhagic fevers in primates in the event of prolonged contact without Animal studies have given rise to divergent results. One team
any barrier precautions. It was also discussed in 1970 during an succeeded in triggering both a cell-mediated and a humoral
outbreak of Lassa fever in Nigeria. Whatever the case, it would immune response in mice using a vaccine strain, but this did not
probably only take place during the most advanced stages of the reduce the fatality rate [59]. By using two doses of vaccine, the same
disease [44]. team then demonstrated reduced mortality in a mouse model [60].
P. Fillâtre et al. / Médecine et maladies infectieuses 49 (2019) 574–585 579

Phase 1 vaccine trials are currently underway in humans, but the Table 2
Clinical signs reported in the literature.
antibodies produced only seem to have a limited virus neutraliza-
Signes cliniques rapportés dans la littérature.
tion capability [61].
Reported References
frequency, %
9. Clinical presentation
Common clinical signs
Pre-hemorrhagic phase
The largest series of clinical reports are from highly endemic Fever 43–98 [63,64]
countries (Iran and Turkey) and should be considered with caution Myalgia 54–93 [62,63]
because based on IgM seropositivity [31,62–65] rather than direct Headache 48–80 [31,65]
Nausea/vomiting 46–83 [63,64]
detection of the CCHF virus. Based on the very high seropositiv-
Diarrhea 31–44 [62,65]
ity rates found in some endemic areas, up to 27% in some regions Retro-orbital pain –
of Romania and Greece, a great many cases of CCHF are either Stiff neck –
asymptomatic or cause symptoms so limited that patients do not Hepatomegaly 30–37 [31,62]
Splenomegaly 14–37 [31,65]
seek medical care [66,67]. It is estimated that 88% of seropositive
Peripheral lymphadenopathy 13 [31]
patients in Turkey experienced only limited symptoms [68]. Hemorrhagic phase
The clinical features most commonly reported in the literature, Epistaxis 17–52 [31,63]
which are also the most extreme signs, are detailed in Table 2. The Hematemesis 8–31 [31,64]
mean incubation time is 2 to 7 days. Longer incubation times have Melena 1–20 [31,64]
Hemoptysia 6–9 [62,63,64]
been described in the literature. In a series of 312 patients with
Hematuria 15–37 [62,63,64]
CCHF after a tick bite in Turkey, 12 patients (3.8%) displayed an incu- Ecchymosis/Purpura 20–47 [62,63]
bation time > 12 days which is the generally accepted maximum Rare clinical signs
incubation time (longest: 53 days) [69]. These patients displayed Lungs
Alveolar hemorrhage [150,151]
less severe symptoms (less fever, less cytolysis and lower mortal-
Hemothorax [152,153]
ity). Heart
The disease can be divided into 3 phases, similarly to other viral Left ventricular dysfunction [154]
hemorrhagic fevers (Fig. 3). During the pre-hemorrhagic phase, Myocarditis [156]
symptoms are generally unspecific. The main symptom is high Pericardial effusion [154,157]
Pulmonary arterial hypertension [155,158]
fever, which can reach 39–41 ◦ C and usually lasts 4–5 days [1]. It
Bundle branch block or repolarization [158]
can be associated with myalgia, headache, retro-orbital pain and disturbance
sometimes a stiff neck. Gastrointestinal symptoms then occur with Eyes
nausea and/or vomiting and more rarely diarrhea [31,62–65]. The Retinal and subconjunctival hemorrhage [159]
Skin
pre-hemorrhagic phase is generally short but can last up to one
Erythema nodosum [160]
week [1]. Mobilliform eruption [161]
Infection with the CCHF virus results in endothelial dysfunc- Digestive system
tion and capillary leaking of red blood cells and plasma into tissue. Acalculous cholecystitis [162]
Endothelial damage leads to activation of the coagulation cascade Acute pancreatitis [163]
Ascites [164,165]
and thrombocytopenia, which increases bleeding [22]. The hemor-
Spontaneous retroperitoneal hematoma [166]
rhagic phase is characterized by mucous tissue bleeding: epistaxis, Urinary system
hematemesis, more rarely melena, hemoptysis and hematuria. Orchiepididymitis [167]
Bleeding into the skin (ecchymosis/purpura) is often observed ENT
[31,62–65]. Parotitis [168]
Endocrine system
During the first two phases, the main signs detected upon exam- Disturbed vasopressin secretion [169]
ination are hepatomegaly in approximately one third of cases, Adrenal gland failure [170]
splenomegaly and sometimes peripheral lymph node enlargement Brain
[31,62]. The clinical features associated with patient mortality are Acute subdural hematoma [171]
Kidneys
somnolence, gross hematuria, hematemesis and melena [1,70,71].
Proteinuria and elevated neutrophil [172]
A fatality rate of 5–30% is generally described with death occurring gelatinase-associated lipocalin (NGAL)
during the hemorrhagic phase. A mortality of 80% was reported in Bones and joints
the first series whereas in the most recent last Turkish series the Inflammatory arthralgia [173]
mortality rate was 4,6% [1,5].
The convalescence phase starts 10–20 days after the onset of
hepatocellular cytolysis are often observed (Fig. 3). Many studies
the first clinical symptoms [1]. Patients experience marked fatigue,
have demonstrated that the higher the degree of thrombocytope-
tachycardia with labile blood pressure, temporary alopecia and
nia, decreased PT, cytolysis, and longer aPTT and the poorer the
memory impairment. The convalescence phase usually lasts about
prognosis [72,73]. In rare cases, a macrophage activation syndrome
ten days [1].
enhances cytopenia [74–76]. Finally, elevated LDH and CK levels
are frequently observed [1].
10. Laboratory findings
10.2. Imaging features
10.1. Clinical chemistry
Unspecific abnormal features are observed using abdomi-
Thrombocytopenia is observed in almost all cases, as is a nal ultrasound [77,78], and chest [79] or abdominal CT [80].
decreased prothrombin time (PT), increased activated partial Evidence of deep lymph node enlargement, serous effusion, hep-
thromboplastin time (aPTT) and hypofibrinogenemia. These atosplenomegaly and pulmonary infiltration does not change
abnormalities are highest during the hemorrhagic phase before patient management. Systematic imaging provides no real benefit,
slowly regressing [1]. At the same time, transient leukopenia and especially since moving the patient increases the risk of secondary
580 P. Fillâtre et al. / Médecine et maladies infectieuses 49 (2019) 574–585

Fig. 3. Phases of Crimean-Congo hemorrhagic fever, adapted from [1].


Différentes phases de la fièvre hémorragique de Crimée-Congo.

contamination if containment measures are breached. European methods have been described with primers common to all known
guidelines even recommend to perform bedside chest X-rays, ultra- viral strains [22]. The quantitative PCR developed by Drosten [83]
sound with equipment used only in the isolation unit, and to can be used to detect the genomes of the Ebola, Marburg, Lassa,
minimize the use of CT or MRI [54]. CCHF, Rift valley fever, Dengue and yellow fever viruses in a single
assay, which may prove useful for travelers returning from an area
11. Diagnosis where several viral hemorrhagic fevers are found.
The main diagnostic challenge is caused by the very short win-
11.1. Clinical chemistry dow during which the virus can be detected in blood samples using
PCR. If symptomatic patients are not rapidly admitted to hospi-
Patient samples should be considered as highly infectious and tal, PCR results may be negative and serological testing required.
represent a high risk for the laboratory technicians and pathologists But IgM levels can remain high for several months and although
who handle them. Lab investigations should therefore be limited to they demonstrate recent infection, they do not prove that the
the tests strictly necessary to: symptoms are caused by the CCHF virus. The diagnostic value of
seropositivity therefore depends on the geographical region. In an
• confirm diagnosis; endemic area like Turkey, seropositivity may reflect recent CCHF
• determine consequences (impaired hemostasis, electrolyte infection that caused limited symptoms, and the unspecific clin-
imbalance); ical signs that have led the patient to seek care may be due to
• and allow alternative diagnosis. a number of other potential diseases. Such symptoms may be
caused by other hemorrhagic fevers depending on the region vis-
ited, Rickettsial diseases, Q fever, leptospirosis, malaria, bacterial
CCHF virus isolation and culture in order to confirm diagnosis
sepsis, viral hepatitis and infective endocarditis, etc [1,23]. Due
should only be performed in a BSL-4 laboratory. Once inactivated,
to the null prevalence of CCHF in Western Europe, positive spo-
other samples may be handled using BSL-3 biosafety precautions
radic IgM results should however be considered as of diagnostic
[49].
relevance.

11.2. Serology
12. Severity scoring
Specific IgG and IgM antibodies can be detected by ELISA. IgM
antibodies are detected from the 5th to 7th day on [22], and disap- Two different scoring systems have been proposed to assess
pear 4 months after infection. However, specific IgGs are detected prognosis upon admission of patients with CCHF. Both systems are
for at least 5 years [23]. An acute phase infection may therefore be estimated to predict the risk of unfavorable prognosis with a sensi-
documented by detecting IgMs (but not IgGs) in an initial serum tivity of 96–100% and a specificity of 93–100% [84,85] These scores
sample, then IgMs and IgGs at the same time, or seroconversion can help in predicting a patient’s clinical outcome and have been
with a four-fold increase in the antibody titer between two succes- developed for use in endemic countries. The first scoring system
sive samples of serum [6]. The specificity of serological testing is was designed to help decide whether patients required transferring
excellent, with no reported false positives or cross-reactivity with to a reference center or whether they could be treated satisfacto-
other viruses. However, the sensitivity of IgM detection ranges from rily in a local hospital [84]. The second system was developed in an
87.8 to 100% [81,82]. uncontrolled retrospective study to stratify patients based on the
risk of death, the patients at intermediate risk having benefited to
11.3. Quantitative PCR a greater degree from treatment with ribavirin [85]. Although not
included in the clinical scoring systems, a high viral load exceed-
Molecular biology tests have now become the preferred meth- ing 108 copies/mL was also independently associated with poor
ods for fast, exact diagnosis. Various different real-time PCR prognosis [86–88].
P. Fillâtre et al. / Médecine et maladies infectieuses 49 (2019) 574–585 581

13. Treatment options temporary use authorization), WHO recommends 17 mg/kg (max-
imum dose: 1 g) every 6 hours from day 1 to day 4, then 8 mg/kg
13.1. Symptomatic care (maximum dose: 500 mg) every 8 hours from day 5 to day 10 [109].
Based on encouraging retrospective data [47,110,111], some
Care for patients with CCHF is based on symptomatic treatment authors recommend post-exposure treatment with ribavirin for
[49]: correcting hypovolemia and electrolyte imbalance caused by health professionals accidentally exposed to the virus, especially
gastrointestinal disturbances, correcting hemostasis in the event in the event of needlestick injury [48]. A recent meta-analysis
of bleeding (fresh frozen plasma, platelet transfusion), packed cell estimated that such prophylactic post-exposure treatment signifi-
transfusion [1,23]. Potential coinfection should be investigated and cantly reduced the risk of infection (OR 0.01 [0–0.003]) [112].
treated because although rarely described, bacterial coinfection
[89,90] and concomitant malaria [91,92] have been reported in the 13.3. Other treatment options
literature.
The efficacy of favipiravir, a broad-spectrum viral polymerase
13.2. Ribavirin inhibitor was assessed in a mouse model and demonstrated superi-
ority over ribavirin and a persistent effect even when administered
Tests performed on ribavirin, a nucleoside analog, have demon- at a late stage after the onset of symptoms [94]. However, too little
strated that, in vitro, it inhibits CCHF virus replication and its data is currently available to be able to recommend the routine use
cytopathogenic effect in adrenal carcinoma cells [93]. Ribavirin of favipiravir to treat CCHF.
also seems to be able to decrease the mortality rate in a mouse Other therapeutic options have been experimented. Of particu-
model, but only if administered at an early stage of the disease lar interest: plasmapheresis combined with ribavirin in patients
[95]. Treatment with ribavirin is contraindicated in pregnant and with severe CCHF in an attempt to reduce the proinflam-
breast-feeding women, or in patients with pre-existing heart dis- matory cytokine load [113,114], high doses of corticosteroids
ease. (5–30 mg/kg/day) to treat macrophage activation syndrome, poly-
In the first years of the 21st century, ribavirin was therefore valent immunoglobulins and fresh frozen plasma [115].
recommended in various clinical guidelines, notably because ret- Two Iranian studies attempted to evaluate the potential ben-
rospective studies on cases in Turkey and Iran showed a lower efit of immunomodulatory therapy. The first study compared the
mortality rate [62,95], or even rapid recovery [96,97] in the patients combined use of polyvalent immunoglobulins and ribavirin (n = 12)
who received ribavirin, especially when administered at an early with ribavirin alone (n = 28) and showed a beneficial effect of
stage (within 4 days of the onset of symptoms) [98,99]. In a recent polyvalent immunoglobulins in terms of time to recovery from
retrospective study adjusted using a severity score, ribavirin had cytopenia and return to normal liver function. Unfortunately, the
a protective effect against death with an odds ratio (OR) of 0.04 study was not double-blinded [116]. The second trial was an
[0.004–0.48] [85], thus confirming the results of the earlier stud- open, controlled, prospective study comparing the combined use
ies that demonstrated the efficacy of ribavirin for the most severe of ribavirin and 10 mg/kg/d corticosteroids (n = 13) with ribavirin
cases [62]. In the literature review published in 2010 by Soares- alone (n = 22). The corticosteroid group were found to have less
Weiser et al. [101] that included 11 observational studies and a severe hemorrhagic syndrome, a reduced need for transfusion and
total of 955 patients, ribavirin was also shown to have a benefi- recovered faster from cytopenia. However, the mortality rate was
cial effect on mortality, with a risk ratio (RR) of death estimated at equivalent in both groups [117].
0.56 [0.35–0.90]. However, other retrospective studies have shown Another interesting option is worth mentioning even though
a lack of effect [101–104], and some experts are convinced that only preliminary findings are currently available: Kubar et al.
ribavirin is of no clinical value [105], all the more so after Bodur reported interesting results in CCHF patients with a high viral load
et al. reported that the viral load in patients who received oral (n = 15) after administration of hyperimmune globulins prepared
ribavirin (n = 10) was not significantly reduced compared with from the serum of convalescent patients [118].
patients receiving only symptomatic treatment (n = 40) [106].
Studies with a higher level of evidence are rare and suggest a
lack of efficacy for ribavirin. In a randomized trial on the efficacy of 14. Conclusions
ribavirin [107], no differences were found between patients in the
ribavirin + symptomatic care group (n = 64) compared with patients The presence of the ticks that vector CCHF in Western Europe
receiving only symptomatic care (n = 72) in terms of mortality, and the favorable climate make it plausible that indigenous cases
length of hospital stay, need for transfusion and platelet count of CCHF will emerge in France in the future. Further investigation
recovery time. This unblinded study included all patients with the of the contagiousness and severity of the CCHF virus are required
clinical features of hemorrhagic fever suspected to be CCHF. Only to better understand the disease and implement adequate patient
documented cases were included, but the population size required isolation measures in order to prevent the secondary infection of
to achieve a predefined power was not calculated so it is impossible health professionals.
to draw meaningful conclusions from the findings.
Two meta-analyses that assessed the efficacy of the use of rib- Disclosure of interest
avirin to reduce mortality from CCHF concluded that too little data
had been published to determine whether ribavirin has a beneficial The authors declare that they have no competing interest.
effect or not [100,108]. Pending the results of a well-documented
study and because a lack of evidence does not necessarily imply a
References
lack of effect, WHO has based its guidelines on the efficacy demon-
strated in vitro and in animal models, as well as on the retrospective [1] Ergönül O. Crimean-Congo haemorrhagic fever. Lancet Infect Dis
data published and the low number of adverse effects, and recom- 2006;6(4):203–14.
mends that patients with CCHF be given an oral loading dose of 2 g [2] Whitehouse CA. Crimean-Congo hemorrhagic fever. Antiviral Res
2004;64(3):145–60.
of ribavirin, followed by 1 g every 6 hours from day 1 to day 4 of [3] Bente DA, Forrester NL, Watts DM, McAuley AJ, Whitehouse CA, Bray M.
treatment, then 500 mg every 6 hours from day 5 to day 10. If intra- Crimean-Congo hemorrhagic fever: history, epidemiology, pathogenesis,
venous treatment is possible (poor availability and very limited clinical syndrome and genetic diversity. Antiviral Res 2013;100(1):159–89.
582 P. Fillâtre et al. / Médecine et maladies infectieuses 49 (2019) 574–585

[4] Papa A, Weber F, Hewson R, Weidmann M, Koksal I, Korukluoglu G, et al. Meet- [32] Yilmaz GR, Buzgan T, Irmak H, Safran A, Uzun R, Cevik MA, et al. The epidemi-
ing report: First International Conference on Crimean-Congo hemorrhagic ology of Crimean-Congo hemorrhagic fever in Turkey, 2002–2007. Int J Infect
fever. Antiviral Res 2015;120:57–65. Dis 2009;13(3):380–6.
[5] Spengler JR, Bente DA, Bray M, Burt F, Hewson R, Korukluoglu G, et al. Second [33] Estrada-Peña A, Ayllón N, de la Fuente J. Impact of climate trends on tick-borne
International Conference on Crimean-Congo Hemorrhagic Fever. Antiviral Res pathogen transmission. Front Physiol 2012;3:64.
2018;150:137–47. [34] Papa A, Sidira P, Kallia S, Ntouska M, Zotos N, Doumbali E, et al. Factors
[6] Charrel RN, Attoui H, Butenko AM, Clegg JC, Deubel V, Frolova TV, et al. associated with IgG positivity to Crimean-Congo hemorrhagic fever virus
Tick-borne virus diseases of human interest in Europe. Clin Microbiol Infect in the area with the highest seroprevalence in Greece. Ticks Tick-Borne Dis
2004;10(12):1040–55. 2013;4(5):417–20.
[7] Papa A, Christova I, Papadimitriou E, Antoniadis A. Crimean-Congo hemor- [35] Lwande OW, Irura Z, Tigoi C, Chepkorir E, Orindi B, Musila L, et al. Seropreva-
rhagic fever in Bulgaria. Emerg Infect Dis 2004;10(8):1465–7. lence of Crimean Congo hemorrhagic fever virus in Ijara District, Kenya. Vector
[8] Mertens M, Schuster I, Sas MA, Vatansever Z, Hubalek Z, Güven E, et al. Borne Zoonotic Dis Larchmt N 2012;12(9):727–32.
Crimean-Congo Hemorrhagic Fever Virus in Bulgaria and Turkey. Vector [36] Nurmakhanov T, Sansyzbaev Y, Atshabar B, Deryabin P, Kazakov S, Zhol-
Borne Zoonotic Dis Larchmt N 2016;16(9):619–23. shorinov A, et al. Crimean-Congo haemorrhagic fever virus in Kazakhstan
[9] Sas MA, Mertens M, Kadiat JG, Schuster I, Pongombo CPS, Maloba AGK, (1948–2013). Int J Infect Dis 2015;38:19–23.
et al. Serosurvey for Crimean-Congo hemorrhagic fever virus infections in [37] Mostafavi E, Pourhossein B, Esmaeili S, Bagheri Amiri F, Khakifirouz S,
ruminants in Katanga province, Democratic Republic of the Congo. Ticks Tick- Shah-Hosseini N, et al. Seroepidemiology and risk factors of Crimean-Congo
Borne Dis 2017;8(6):858–61. Hemorrhagic Fever among butchers and slaughterhouse workers in south-
[10] Maiga O, Sas MA, Rosenke K, Kamissoko B, Mertens M, Sogoba N, et al. Serosur- eastern Iran. Int J Infect Dis 2017;64:85–9.
vey of Crimean-Congo Hemorrhagic Fever Virus in Cattle, Mali, West Africa. [38] Pshenichnaya NY, Sydenko IS, Klinovaya EP, Romanova EB, Zhuravlev AS. Pos-
Am J Trop Med Hyg 2017;96(6):1341–5. sible sexual transmission of Crimean-Congo hemorrhagic fever. Int J Infect Dis
[11] Sas MA, Mertens M, Isselmou E, Reimer N, El Mamy BO, Doumbia B, et al. 2016;45:109–11.
Crimean-Congo Hemorrhagic Fever Virus-Specific Antibody Detection in Cat- [39] Ergonul O, Celikbas A, Yildirim U, Zenciroglu A, Erdogan D, Ziraman I, et al.
tle in Mauritania. Vector Borne Zoonotic Dis Larchmt N 2017;17(8):582–7. Pregnancy and Crimean-Congo haemorrhagic fever. Clin Microbiol Infect
[12] Suliman HM, Adam IA, Saeed SI, Abdelaziz SA, Haroun EM, Aradaib IE. 2010;16(6):647–50.
Crimean Congo hemorrhagic fever among the one-humped camel (Camelus [40] Aydemir O, Erdeve O, Oguz SS, Dilmen U. A healthy newborn born to a mother
dromedaries) in Central Sudan. Virol J 2017;14(1):147. with Crimean-Congo hemorrhagic fever: is there protection from transpla-
[13] Spengler JR, Bergeron É, Rollin PE. Seroepidemiological Studies of Crimean- cental transmission? Int J Infect Dis 2010;14(5):e450.
Congo Hemorrhagic Fever Virus in Domestic and Wild Animals. PLoS Negl [41] Green ST. Bad bugs travel as well as happy holidaymakers. BMJ
Trop Dis 2016;10(1):e0004210. 2002;325(7369):905.
[14] Papa A, Velo E, Papadimitriou E, Cahani G, Kota M, Bino S. Ecology of the [42] Athar MN, Baqai HZ, Ahmad M, Khalid MA, Bashir N, Ahmad AM, et al. Short
Crimean-Congo hemorrhagic fever endemic area in Albania. Vector Borne report: Crimean-Congo hemorrhagic fever outbreak in Rawalpindi, Pakistan,
Zoonotic Dis Larchmt N 2009;9(6):713–6. February 2002. Am J Trop Med Hyg 2003;69(3):284–7.
[15] Mancini F, Toma L, Ciervo A, Di Luca M, Faggioni G, Lista F, et al. Virus investiga- [43] Harxhi A, Pilaca A, Delia Z, Pano K, Rezza G. Crimean-Congo hemorrhagic
tion in ticks from migratory birds in Italy. New Microbiol 2013;36(4):433–4. fever: a case of nosocomial transmission. Infection 2005;33(4):295–6.
[16] Chitimia-Dobler L, Nava S, Bestehorn M, Dobler G, Wölfel S. First detection of [44] Weber DJ, Rutala WA. Risks and prevention of nosocomial transmission of
Hyalomma rufipes in Germany. Ticks Tick-Borne Dis 2016;7(6):1135–8. rare zoonotic diseases. Clin Infect Dis 2001;32(3):446–56.
[17] Vial L, Stachurski F, Leblond A, Huber K, Vourc’h G, René-Martellet M, et al. [45] Smego RA, Sarwari AR, Siddiqui AR. Crimean-Congo hemorrhagic fever: pre-
Strong evidence for the presence of the tick Hyalomma marginatum Koch, vention and control limitations in a resource-poor country. Clin Infect Dis
1844 in southern continental France. Ticks Tick-Borne Dis 2016;7(6):1162–7. 2004;38(12):1731–5.
[18] Palomar AM, Portillo A, Santibáñez S, García-Álvarez L, Muñoz-Sanz A, [46] Naderi H, Sheybani F, Bojdi A, Khosravi N, Mostafavi I. Fatal nosocomial spread
Márquez FJ, et al. Molecular (ticks) and serological (humans) study of of Crimean-Congo hemorrhagic fever with very short incubation period. Am
Crimean-Congo hemorrhagic fever virus in the Iberian Peninsula, 2013-2015. J Trop Med Hyg 2013;88(3):469–71.
Enferm Infecc Microbiol Clin 2017;35(6):344–7. [47] Leblebicioglu H, Sunbul M, Guner R, Bodur H, Bulut C, Duygu F,
[19] Estrada-Peña A, Palomar AM, Santibáñez P, Sánchez N, Habela MA, Portillo A, et al. Healthcare-associated Crimean-Congo haemorrhagic fever in Turkey,
et al. Crimean-Congo hemorrhagic fever virus in ticks, Southwestern Europe, 2002–2014: a multicentre retrospective cross-sectional study. Clin Microbiol
2010. Emerg Infect Dis 2012;18(1):179–80. Infect 2016;22(4):387.e1–4.
[20] Palomar AM, Portillo A, Santibáñez P, Mazuelas D, Arizaga J, Crespo A, [48] Ergonul O. Evidence supports ribavirin use in Crimean-Congo hemorrhagic
et al. Crimean-Congo hemorrhagic fever virus in ticks from migratory birds, fever. Int J Infect Dis 2014;29:296.
Morocco. Emerg Infect Dis 2013;19(2):260–3. [49] HCSP. Infections virales aiguës, importées, hautement contagieuses, et
[21] Gargili A, Estrada-Peña A, Spengler JR, Lukashev A, Nuttall PA, Bente DA. The leur prise en charge [Internet]; 2001 [cited 2018 Feb 23; Available from:
role of ticks in the maintenance and transmission of Crimean-Congo hemor- https://www.hcsp.fr/Explore.cgi/Ouvrage?clef=70].
rhagic fever virus: a review of published field and laboratory studies. Antiviral [50] Pshenichnaya NY, Nenadskaya SA. Probable Crimean-Congo hemorrhagic
Res 2017;144:93–119. fever virus transmission occurred after aerosol-generating medical proce-
[22] Papa A, Mirazimi A, Köksal I, Estrada-Pena A, Feldmann H. Recent advances in dures in Russia: nosocomial cluster. Int J Infect Dis 2015;33:120–2.
research on Crimean-Congo hemorrhagic fever. J Clin Virol 2015;64:137–43. [51] Leblebicioglu H, Ozaras R, Fletcher TE, Beeching NJ, ESCMID Study Group
[23] Leblebicioglu H, Ozaras R, Irmak H, Sencan I. Crimean-Congo hemor- for Infections in Travellers and Migrants (ESGITM). Crimean-Congo haem-
rhagic fever in Turkey: current status and future challenges. Antiviral Res orrhagic fever in travellers: a systematic review. Travel Med Infect Dis
2016;126:21–34. 2016;14(2):73–80.
[24] Al-Abri SS, Abaidani IA, Fazlalipour M, Mostafavi E, Leblebicioglu H, Pshenich- [52] Jauréguiberry S, Tattevin P, Tarantola A, Legay F, Tall A, Nabeth P,
naya N, et al. Current status of Crimean-Congo haemorrhagic fever in the et al. Imported Crimean-Congo hemorrhagic Fever. J Clin Microbiol
World Health Organization Eastern Mediterranean Region: issues, challenges, 2005;43(9):4905–7.
and future directions. Int J Infect Dis 2017;58:82–9. [53] COREB national. Fièvres Hémorragiques Virales (FHV) : repérer et pren-
[25] Gale P, Estrada-Peña A, Martinez M, Ulrich RG, Wilson A, Capelli G, et al. The dre en charge un patient suspect en France. http://www.infectiologie.
feasibility of developing a risk assessment for the impact of climate change on com/UserFiles/File/coreb/20180803-reperer-prendre-en-charge-fhv-ebolard
the emergence of Crimean-Congo haemorrhagic fever in livestock in Europe: c-3aout2018final.pdf.
a review. J Appl Microbiol 2010;108(6):1859–70. [54] Brouqui P, Puro V, Fusco FM, Bannister B, Schilling S, Follin P, et al. Infec-
[26] Maltezou HC, Papa A. Crimean-Congo hemorrhagic fever: risk for emergence tion control in the management of highly pathogenic infectious diseases:
of new endemic foci in Europe? Travel Med Infect Dis 2010;8(3):139–43. consensus of the European Network of Infectious Disease. Lancet Infect Dis
[27] Dreshaj S, Ahmeti S, Ramadani N, Dreshaj G, Humolli I, Dedushaj I. Current 2009;9(5):301–11.
situation of Crimean-Congo hemorrhagic fever in Southeastern Europe and [55] Gürbüz Y, Sencan I, Oztürk B, Tütüncü E. A case of nosocomial transmission
neighboring countries: a public health risk for the European Union? Travel of Crimean-Congo hemorrhagic fever from patient to patient. Int J Infect Dis
Med Infect Dis 2016;14(2):81–91. 2009;13(3):e105–7.
[28] Negredo A, de la Calle-Prieto F, Palencia-Herrejón E, Mora-Rillo M, [56] Yildirmak T, Tulek N, Bulut C. Crimean-Congo haemorrhagic fever: transmis-
Astray-Mochales J, Sánchez-Seco MP, et al. Autochthonous Crimean-Congo sion to visitors and healthcare workers. Infection 2016;44(5):687–9.
Hemorrhagic Fever in Spain. N Engl J Med 2017 13;377(2):154–61. [57] Ergonul O, Zeller H, Celikbas A, Dokuzoguz B. The lack of Crimean-Congo hem-
[29] ProMED-mail - Archive Number: 20180811.5957008 [Internet]. [cited 2018 orrhagic fever virus antibodies in healthcare workers in an endemic region.
Sep 5]. Available from: https://www.promedmail.org/. Int J Infect Dis 2007;11(1):48–51.
[30] Santé Publique France. Définition des priorités dans le domaine des zoonoses [58] Tarantola A, Nabeth P, Tattevin P, Michelet C, Zeller H, Incident Management
non alimentaires; 2008–2009 [Internet; cited 2018 Feb 23; Available from: Group. Lookback exercise with imported Crimean-Congo hemorrhagic fever,
http://www.invs.santepubliquefrance.fr/pmb/invs/(id)/PMB503]. Senegal and France. Emerg Infect Dis 2006;12(9):1424–6.
[31] Bakir M, Ugurlu M, Dokuzoguz B, Bodur H, Tasyaran MA, Vahaboglu H, et al. [59] Dowall SD, Buttigieg KR, Findlay-Wilson SJD, Rayner E, Pearson G,
Crimean-Congo haemorrhagic fever outbreak in Middle Anatolia: a multi- Miloszewska A, et al. A Crimean-Congo hemorrhagic fever (CCHF) viral vac-
centre study of clinical features and outcome measures. J Med Microbiol cine expressing nucleoprotein is immunogenic but fails to confer protection
2005;54(Pt 4):385–9. against lethal disease. Hum Vaccines Immunother 2016;12(2):519–27.
P. Fillâtre et al. / Médecine et maladies infectieuses 49 (2019) 574–585 583

[60] Buttigieg KR, Dowall SD, Findlay-Wilson S, Miloszewska A, Rayner E, Hewson [86] Cevik MA, Erbay A, Bodur H, Eren SS, Akinci E, Sener K, et al. Viral load as
R, et al. A novel vaccine against Crimean-Congo Haemorrhagic Fever pro- a predictor of outcome in Crimean-Congo hemorrhagic fever. Clin Infect Dis
tects 100% of animals against lethal challenge in a mouse model. PloS One 2007;45(7):e96–100.
2014;9(3):e91516. [87] Duh D, Saksida A, Petrovec M, Ahmeti S, Dedushaj I, Panning M, et al. Viral
[61] Mousavi-Jazi M, Karlberg H, Papa A, Christova I, Mirazimi A. Healthy individ- load as predictor of Crimean-Congo hemorrhagic fever outcome. Emerg Infect
uals’ immune response to the Bulgarian Crimean-Congo hemorrhagic fever Dis 2007;13(11):1769–72.
virus vaccine. Vaccine 2012;30(44):62259. [88] Hasanoglu I, Guner R, Carhan A, Kocak Tufan Z, Yagci-Caglayik D, Guven T,
[62] Ergönül O, Celikbaş A, Dokuzoguz B, Eren S, Baykam N, Esener H. Char- et al. Crucial parameter of the outcome in Crimean Congo hemorrhagic fever:
acteristics of patients with Crimean-Congo hemorrhagic fever in a recent Viral load. J Clin Virol 2016;75:42–6.
outbreak in Turkey and impact of oral ribavirin therapy. Clin Infect Dis 2004; [89] Sunbul M, Leblebicioglu H, Fletcher TE, Elaldi N, Ozkurt Z, Bastug A, et al.
39(2):284–7. Crimean-Congo haemorrhagic fever and secondary bacteraemia in Turkey. J
[63] Alavi-Naini R, Moghtaderi A, Koohpayeh H-R, Sharifi-Mood B, Naderi M, Infect 2015;71(5):597–9.
Metanat M, et al. Crimean-Congo hemorrhagic fever in Southeast of Iran. J [90] Duygu F, Sari T, Kaya T, Bulut N. Brucellosis in patients with Crimean-Congo
Infect 2006;52(5):378–82. hemorrhagic fever. J Arthropod-Borne Dis 2017;11(4):463–8.
[64] Ergonul O, Celikbas A, Baykam N, Eren S, Dokuzoguz B. Analysis of risk-factors [91] Sharifi-Mood B, Metanat M, Rakhshani F, Shakeri A. Co-infection of malaria
among patients with Crimean-Congo haemorrhagic fever virus infection: and Crimean-Congo hemorrhagic fever. Iran J Parasitol 2011;6(3):113–5.
severity criteria revisited. Clin Microbiol Infect 2006;12(6):551–4. [92] Christova I, Petrov A, Papa A, Vutchev D, Kalvatchev N, Vatev N, et al. Fatal
[65] Kilinc C, Gückan R, Capraz M, Varol K, Zengin E, Mengeloglu Z, et al. outcome of coinfection of Crimean-Congo hemorrhagic fever and malaria. Jpn
Examination of the specific clinical symptoms and laboratory findings of J Infect Dis 2015;68(2):131–4.
Crimean-Congo hemorrhagic fever. J Vector Borne Dis 2016;53(2):162–7. [93] Paragas J, Whitehouse CA, Endy TP, Bray M. A simple assay for determining
[66] Sidira P, Maltezou HC, Haidich A-B, Papa A. Seroepidemiological study of antiviral activity against Crimean-Congo hemorrhagic fever virus. Antiviral
Crimean-Congo haemorrhagic fever in Greece, 2009–2010. Clin Microbiol Res 2004;62(1):21–5.
Infect 2012;18(2):E16–9. [94] Hawman DW, Haddock E, Meade-White K, Williamson B, Hanley PW, Rosenke
[67] Ceianu CS, Panculescu-Gatej RI, Coudrier D, Bouloy M. First serologic evidence K, et al. Favipiravir (T-705) but not ribavirin is effective against two dis-
for the circulation of Crimean-Congo hemorrhagic fever virus in Romania. tinct strains of Crimean-Congo hemorrhagic fever virus in mice. Antiviral Res
Vector Borne Zoonotic Dis Larchmt N 2012;12(9):718–21. 2018;157:18–26.
[68] Bodur H, Akinci E, Ascioglu S, Öngürü P, Uyar Y. Subclinical infections [95] Mardani M, Jahromi MK, Naieni KH, Zeinali M. The efficacy of oral ribavirin
with Crimean-Congo hemorrhagic fever virus, Turkey. Emerg Infect Dis in the treatment of crimean-congo hemorrhagic fever in Iran. Clin Infect Dis
2012;18(4):640–2. 2003;36(12):1613–8.
[69] Kaya A, Engin A, Güven AS, Içağasıoğlu FD, Cevit O, Elaldi N, et al. Crimean- [96] Sharifi-Mood B, Metanat M, Ghorbani-Vaghei A, Fayyaz-Jahani F, Akrami E.
Congo hemorrhagic fever disease due to tick bite with very long incubation The outcome of patients with Crimean-Congo hemorrhagic fever in Zahedan,
periods. Int J Infect Dis 2011;15(7):e449–52. southeast of Iran: a comparative study. Arch Iran Med 2009;12(2):151–3.
[70] Hatipoglu CA, Bulut C, Yetkin MA, Ertem GT, Erdinc FS, Kilic EK, et al. [97] Ozkurt Z, Kiki I, Erol S, Erdem F, Yilmaz N, Parlak M, et al. Crimean-Congo
Evaluation of clinical and laboratory predictors of fatality in patients with hemorrhagic fever in Eastern Turkey: clinical features, risk factors and efficacy
Crimean-Congo haemorrhagic fever in a tertiary care hospital in Turkey. of ribavirin therapy. J Infect 2006;52(3):207–15.
Scand J Infect Dis 2010;42(6–7):516–21. [98] Izadi S, Salehi M. Evaluation of the efficacy of ribavirin therapy on survival of
[71] Cevik MA, Erbay A, Bodur H, Gülderen E, Baştuğ A, Kubar A, et al. Clinical Crimean-Congo hemorrhagic fever patients: a case-control study. Jpn J Infect
and laboratory features of Crimean-Congo hemorrhagic fever: predictors of Dis 2009;62(1):11–5.
fatality. Int J Infect Dis 2008;12(4):374–9. [99] Tasdelen Fisgin N, Ergonul O, Doganci L, Tulek N. The role of ribavirin in the
[72] Tasdelen Fisgin N, Tanyel E, Doganci L, Tulek N. Risk factors for fatal- therapy of Crimean-Congo hemorrhagic fever: early use is promising. Eur J
ity in patients with Crimean-Congo haemorrhagic fever. Trop Doct Clin Microbiol Infect Dis 2009;28(8):929–33.
2009;39(3):158–60. [100] Soares-Weiser K, Thomas S, Thomson G, Garner P. Ribavirin for Crimean-
[73] Ozturk B, Tutuncu E, Kuscu F, Gurbuz Y, Sencan I, Tuzun H. Evaluation of Congo hemorrhagic fever: systematic review and meta-analysis. BMC Infect
factors predictive of the prognosis in Crimean-Congo hemorrhagic fever: new Dis 2010;10:207.
suggestions. Int J Infect Dis 2012;16(2):e89–93. [101] Elaldi N, Bodur H, Ascioglu S, Celikbas A, Ozkurt Z, Vahaboglu H, et al. Efficacy
[74] Karti SS, Odabasi Z, Korten V, Yilmaz M, Sonmez M, Caylan R, et al. Crimean- of oral ribavirin treatment in Crimean-Congo haemorrhagic fever: a quasi-
Congo hemorrhagic fever in Turkey. Emerg Infect Dis 2004;10(8):1379–84. experimental study from Turkey. J Infect 2009;58(3):238–44.
[75] Tasdelen Fisgin N, Fisgin T, Tanyel E, Doganci L, Tulek N, Guler N, et al. [102] Yilmaz R, Kundak AA, Ozer S, Esmeray H. Successful treatment of severe
Crimean-Congo hemorrhagic fever: five patients with hemophagocytic syn- Crimean-Congo hemorrhagic fever with supportive measures without rib-
drome. Am J Hematol 2008;83(1):73–6. avirin and hypothermia. J Clin Virol 2009;44(2):181–2.
[76] Cagatay A, Kapmaz M, Karadeniz A, Basaran S, Yenerel M, Yavuz S, et al. [103] Ertem G, Sönmezer MÇ, Temoçin F, Ataman Hatipoğlu Ç, Tülek N, Oral B. The
Haemophagocytosis in a patient with Crimean Congo haemorrhagic fever. efficacy of oral ribavirin on clinical and laboratory parameters inCrimean-
J Med Microbiol 2007;56(Pt 8):1126–8. Congo hemorrhagic fever: an observational study from Turkey. Turk J Med
[77] Tufan ZK, Yigit H, Kacar M, Bulut C, Canpolat G, Hatipoglu CA, et al. Sci 2016;46(5):1407–14.
Sonographic findings in patients with Crimean-Congo hemorrhagic fever. J [104] Kalın G, Metan G, Demiraslan H, Doganay M. Do we really need rib-
Ultrasound Med 2014;33(11):1999–2003. avirin in the treatment of crimean-congo hemorrhagic fever? J Chemother
[78] Ziraman I, Celikbas A, Ergonul O, Degirmenci T, Uyanik SA, Koparal S, et al. 2014;26(3):146–9.
Crimean-Congo hemorrhagic fever: aid of abdominal ultrasonography in pre- [105] Ceylan B, Calıca A, Ak O, Akkoyunlu Y, Turhan V. Ribavirin is not effective
diction of severity. Vector Borne Zoonotic Dis Larchmt N 2014;14(11):817–20. against Crimean-Congo hemorrhagic fever: observations from the Turkish
[79] Aktaş T, Aktaş F, Özmen Z, Altunkaş A, Kaya T, Demir O. Thorax CT find- experience. Int J Infect Dis 2013;17(10):e799–801.
ings in patients with Crimean-Congo hemorrhagic fever (CCHF). SpringerPlus [106] Bodur H, Erbay A, Akıncı E, Öngürü P, Bayazıt N, Eren SS, et al. Effect of oral
2016;5(1):1823. ribavirin treatment on the viral load and disease progression in Crimean-
[80] Özmen Z, Albayrak E, Özmen ZC, Aktaş F, Aktas T, Duygu F. The evaluation of Congo hemorrhagic fever. Int J Infect Dis 2011;15(1):e44–7.
abdominal findings in Crimean-Congo hemorrhagic fever. Abdom Radiol N Y [107] Koksal I, Yilmaz G, Aksoy F, Aydin H, Yavuz I, Iskender S, et al. The efficacy
2016;41(2):384–90. of ribavirin in the treatment of Crimean-Congo hemorrhagic fever in Eastern
[81] Vanhomwegen J, Alves MJ, Zupanc TA, Bino S, Chinikar S, Karlberg H, et al. Black Sea region in Turkey. J Clin Virol 2010;47(1):65–8.
Diagnostic assays for Crimean-Congo hemorrhagic fever. Emerg Infect Dis [108] Ascioglu S, Leblebicioglu H, Vahaboglu H, Chan KA. Ribavirin for patients with
2012;18(12):1958–65. Crimean-Congo haemorrhagic fever: a systematic review and meta-analysis.
[82] Emmerich P, Mika A, von Possel R, Rackow A, Liu Y, Schmitz H, et al. Sensitive J Antimicrob Chemother 2011;66(6):1215–22.
and specific detection of Crimean-Congo Hemorrhagic Fever Virus (CCHFV)- [109] Roth DC. Application for inclusion of Ribavirin in the WHO model List of
Specific IgM and IgG antibodies in human sera using recombinant CCHFV essential medicines. http://www.archives.who.int/eml/expcom/expcom15/
nucleoprotein as antigen in ␮-capture and IgG immune complex (IC) ELISA applications/newmed/ribaravin/ribavirin.pdf.
tests. PLoS Negl Trop Dis 2018;12(3):e0006366. [110] Celikbas AK, Dokuzoğuz B, Baykam N, Gok SE, Eroğlu MN, Midilli K, et al.
[83] Drosten C, Göttig S, Schilling S, Asper M, Panning M, Schmitz H, et al. Rapid Crimean-Congo hemorrhagic fever among health care workers, Turkey.
detection and quantification of RNA of Ebola and Marburg viruses, Lassa virus, Emerg Infect Dis 2014;20(3):477–9.
Crimean-Congo hemorrhagic fever virus, Rift Valley fever virus, dengue virus, [111] Guner R, Hasanoglu I, Tasyaran MA, Yapar D, Keske S, Guven T, et al. Is rib-
and yellow fever virus by real-time reverse transcription-PCR. J Clin Microbiol avirin prophylaxis effective for nosocomial transmission of Crimean-Congo
2002;40(7):2323–30. hemorrhagic fever? Vector Borne Zoonotic Dis Larchmt N 2014;14(8):601–5.
[84] Bakır M, Gözel MG, Köksal I, Aşık Z, Günal Ö, Yılmaz H, et al. Validation of a [112] Ergönül Ö, Keske Ş, Çeldir MG, Kara İA, Pshenichnaya N, Abuova G,
severity grading score (SGS) system for predicting the course of disease and et al. Systematic review and meta-analysis of postexposure prophylaxis for
mortality in patients with Crimean-Congo hemorrhagic fever (CCHF). Eur J Crimean-Congo hemorrhagic fever virus among healthcare workers. Emerg
Clin Microbiol Infect Dis 2015;34(2):325–30. Infect Dis 2018;24(9):1642–8.
[85] Dokuzoguz B, Celikbas AK, Gök ŞE, Baykam N, Eroglu MN, Ergönül Ö. Sever- [113] Kurnaz F, Metan G, Coskun R, Kaynar L, Eser B, Doganay M. A case of
ity scoring index for Crimean-Congo hemorrhagic fever and the impact of Crimean-Congo haemorrhagic fever successfully treated with therapeutic
ribavirin and corticosteroids on fatality. Clin Infect Dis 2013;57(9):1270–4. plasma exchange and ribavirin. Trop Doct 2011;41(3):181–2.
584 P. Fillâtre et al. / Médecine et maladies infectieuses 49 (2019) 574–585

[114] Meço BC, Memikoğlu O, Ilhan O, Ayyıldız E, Gunt C, Unal N, et al. Double filtra- [141] Barr DA, Aitken C, Bell DJ, Brown CS, Cropley I, Dawood N, et al. First con-
tion plasmapheresis for a case of Crimean-Congo hemorrhagic fever. Transfus firmed case of Crimean-Congo haemorrhagic fever in the UK. Lancet Lond
Apher Sci 2013;48(3):331–4. Engl 2013;382(9902):1458.
[115] Erduran E, Bahadir A, Palanci N, Gedik Y. The treatment of crimean- [142] Burnett MW. Crimean-Congo Hemorrhagic Fever. J Spec Oper Med Peer Rev J
congo hemorrhagic fever with high-dose methylprednisolone, intravenous SOF Med Prof 2015;15(4):96–8.
immunoglobulin, and fresh frozen plasma. J Pediatr Hematol Oncol [143] Mishra AC, Mehta M, Mourya DT, Gandhi S. Crimean-Congo haemorrhagic
2013;35(1):e19–24. fever in India. Lancet Lond Engl 2011;378(9788):372.
[116] Salehi H, Salehi M, Adibi N, Salehi M. Comparative study between Ribavirin [144] Dunster L, Dunster M, Ofula V, Beti D, Kazooba-Voskamp F, Burt F, et al. First
and Ribavirin plus Intravenous Immunoglobulin against Crimean Congo hem- documentation of human Crimean-Congo hemorrhagic fever, Kenya. Emerg
orrhagic fever. J Res Med Sci 2013;18(6):497–500. Infect Dis 2002;8(9):1005–6.
[117] Sharifi-Mood B, Alavi-Naini R, Metanat M, Mohammadi M, Shakeri A, [145] Nabeth P, Thior M, Faye O, Simon F. Human Crimean-Congo hemorrhagic
Amjadi A. Efficacy of high-dose methylprednisolone in patients with fever, Sénégal. Emerg Infect Dis 2004;10(10):1881–2.
Crimean-Congo haemorrhagic fever and severe thrombocytopenia. Trop Doct [146] Nabeth P, Cheikh DO, Lo B, Faye O, Vall IOM, Niang M, et al. Crimean-
2013;43(2):49–53. Congo hemorrhagic fever, Mauritania. Emerg Infect Dis 2004;10(12):
[118] Kubar A, Haciomeroglu M, Ozkul A, Bagriacik U, Akinci E, Sener K, et al. Prompt 2143–9.
administration of Crimean-Congo hemorrhagic fever (CCHF) virus hyperim- [147] Aradaib IE, Erickson BR, Mustafa ME, Khristova ML, Saeed NS, Elageb RM, et al.
munoglobulin in patients diagnosed with CCHF and viral load monitorization Nosocomial outbreak of Crimean-Congo hemorrhagic fever, Sudan. Emerg
by reverse transcriptase-PCR. Jpn J Infect Dis 2011;64(5):439–43. Infect Dis 2010;16(5):837–9.
[119] WHO | Crimean-Congo haemorrhagic fever (CCHF) [Internet]. WHO. [cited [148] Aradaib IE, Erickson BR, Karsany MS, Khristova ML, Elageb RM, Mohamed
2018 Feb 23; Available from: http://www.who.int/emergencies/diseases/ MEH, et al. Multiple Crimean-Congo hemorrhagic fever virus strains are
crimean-congo-haemorrhagic-fever/en/]. associated with disease outbreaks in Sudan, 2008–2009. PLoS Negl Trop Dis
[120] Das P. Infectious diseases surveillance update. Lancet Infect Dis 2001;1(1):7. 2011;5(5):e1159.
[121] Papa A, Bino S, Llagami A, Brahimaj B, Papadimitriou E, Pavlidou V, et al. [149] Balinandi S, Patel K, Ojwang J, Kyondo J, Mulei S, Tumusiime A,
Crimean-Congo hemorrhagic fever in Albania, 2001. Eur J Clin Microbiol Infect et al. Investigation of an isolated case of human Crimean-Congo hem-
Dis 2002;21(8):603–6. orrhagic fever in Central Uganda, 2015. Int J Infect Dis 2018;68:88–93,
[122] Christova I, Di Caro A, Papa A, Castilletti C, Andonova L, Kalvatchev N, et al. http://dx.doi.org/10.1016/j.ijid.2018.01.013. Epub 2018 Jan 31.
Crimean-Congo hemorrhagic fever, southwestern Bulgaria. Emerg Infect Dis [150] Doganci L, Ceyhan M, Tasdeler NF, Sarikayalar H, Tulek N. Crimean
2009;15(6):983–5. Congo hemorrhagic fever and diffuse alveolar haemorrhage. Trop Doct
[123] Lumley S, Atkinson B, Dowall S, Pitman J, Staplehurst S, Busuttil J, et al. Non- 2008;38(4):252–4.
fatal case of Crimean-Congo haemorrhagic fever imported into the United [151] Dogan OT, Engin A, Salk I, Epozturk K, Eren SH, Elaldi N, et al. Evaluation of
Kingdom (ex Bulgaria), June 2014. Euro Surveill Bull Eur Sur Mal Transm Eur respiratory findings in Crimean-Congo hemorrhagic fever. Southeast Asian J
Commun Dis Bull 2014;19(30). Trop Med Public Health 2011;42(5):1100–5.
[124] Papa A, Dalla V, Papadimitriou E, Kartalis GN, Antoniadis A. Emergence [152] Tanir G, Tuygun N, Balaban I, Doksöz O. A case of Crimean-Congo hemorrhagic
of Crimean-Congo haemorrhagic fever in Greece. Clin Microbiol Infect fever with pleural effusion. Jpn J Infect Dis 2009;62(1):70–2.
2010;16(7):843–7. [153] Sahin IO, Güven AS, Kaya A, Güney C, Cevit O, Arslan M. A child with an
[125] Zakhashvili K, Tsertsvadze N, Chikviladze T, Jghenti E, Bekaia M, Kuchuloria unusual complication of Crimean-Congo hemorrhagic fever: hemorrhagic
T, et al. Crimean-Congo hemorrhagic fever in man, Republic of Georgia, 2009. pleural effusion. J Vector Borne Dis 2016;53(1):87–9.
Emerg Infect Dis 2010;16(8):1326–8. [154] Engin A, Yilmaz MB, Elaldi N, Erdem A, Yalta K, Tandogan I, et al. Crimean-
[126] Kuchuloria T, Imnadze P, Chokheli M, Tsertsvadze T, Endeladze M, Mshvi- Congo hemorrhagic fever: does it involve the heart? Int J Infect Dis
dobadze K, et al. Viral hemorrhagic fever cases in the country of Georgia: 2009;13(3):369–73.
Acute Febrile Illness Surveillance Study results. Am J Trop Med Hyg [155] Gülhan B, Kanık-Yüksek S, Çetin İİ, Özkaya-Parlakay A, Tezer H. Myocarditis
2014;91(2):246–8. in a child with Crimean-Congo hemorrhagic fever. Vector Borne Zoonotic Dis
[127] Mamuchishvili N, Salyer SJ, Stauffer K, Geleishvili M, Zakhashvili K, Morgan Larchmt N 2015;15(9):565–7.
J, et al. Notes from the field: Increase in reported Crimean-Congo hemor- [156] Gul I, Kaya A, Güven AS, Karapinar H, Küçükdurmaz Z, Yilmaz A, et al. Cardiac
rhagic fever cases—country of Georgia, 2014. MMWR Morb Mortal Wkly Rep findings in children with Crimean-Congo hemorrhagic fever. Med Sci Monit
2015;64(8):228–9. Int Med J Exp Clin Res 2011;17(8):CR457–60.
[128] Gozalan A, Esen B, Fitzner J, Tapar FS, Ozkan AP, Georges-Courbot M-C, [157] Aktaş T, Aktaş F, Özmen Z, Kaya T. Does Crimean-Congo hemorrhagic fever
et al. Crimean-Congo haemorrhagic fever cases in Turkey. Scand J Infect Dis cause a vasculitic reaction with pulmonary artery enlargement and acute
2007;39(4):332–6. pulmonary hypertension? Lung 2016;194(5):807–12.
[129] Midilli K, Gargili A, Ergonul O, Sengöz G, Ozturk R, Bakar M, et al. [158] Yilmaz MB, Engin A, Bektasoglu G, Zorlu A, Ege MR, Bakir M, et al. Does electro-
Imported Crimean-Congo hemorrhagic fever cases in Istanbul. BMC Infect cardiography at admission predict outcome in Crimean-Congo hemorrhagic
Dis 2007;7:54. fever? J Vector Borne Dis 2011;48(3):150–4.
[130] Jabbari A, Besharat S, Abbasi A, Moradi A, Kalavi K. Crimean-Congo hemor- [159] Engin A, Erdogan H, Ozec AV, Elaldi N, Toker MI, Bakir M, et al. Ocular find-
rhagic fever: case series from a medical center in Golestan province, Northeast ings in patients with Crimean-Congo hemorrhagic fever. Am J Ophthalmol
of Iran (2004). Indian J Med Sci 2006;60(8):327–9. 2009;147(4) [634–8.e1].
[131] Sharifi-Mood B, Mardani M, Keshtkar-Jahromi M, Rahnavardi M, Hatami H, [160] Bijani B, Mardani M, Toosi P. Erythema nodosum in the course of Crimean-
Metanat M. Clinical and epidemiologic features of Crimean-Congo hemor- Congo haemorrhagic fever. Trop Doct 2010;40(2):123–4.
rhagic fever among children and adolescents from southeastern Iran. Pediatr [161] Akyol M, Ozçelik S, Engin A, Hayta SB, Biçici F. Cutaneous manifesta-
Infect Dis J 2008;27(6):561–3. tions of Crimean-Congo haemorrhagic fever: morbilliform eruptions may
[132] Chinikar S, Goya MM, Shirzadi MR, Ghiasi SM, Mirahmadi R, Haeri A, et al. reflect a favorable outcome and not low platelet levels. Eur J Dermatol
Surveillance and laboratory detection system of Crimean-Congo haemor- 2010;20(4):523–4.
rhagic fever in Iran. Transbound Emerg Dis 2008;55(5–6):200–4. [162] Guner R, Hasanoglu I, Yapar D, Tasyaran MA. A case of Crimean Congo hem-
[133] Majeed B, Dicker R, Nawar A, Badri S, Noah A, Muslem H. Morbidity and orrhagic fever complicated with acalculous cholecystitis and intraabdominal
mortality of Crimean-Congo hemorrhagic fever in Iraq: cases reported to abscess. J Clin Virol 2011;50(2):162–3.
the National Surveillance System, 1990–2010. Trans R Soc Trop Med Hyg [163] Bastug A, Kayaaslan B, But A, Aslaner H, Sertcelik A, Akinci E,
2012;106(8):480–3. et al. A case of Crimean-Congo hemorrhagic fever complicated with
[134] Al-Zadjali M, Al-Hashim H, Al-Ghilani M, Balkhiar A. A case of crimean-congo acute pancreatitis. Vector Borne Zoonotic Dis Larchmt N 2014;14(11):
hemorrhagic Fever in Oman. Oman Med J 2013;28(3):210–2. 827–9.
[135] Mohamed Al Dabal L, Rahimi Shahmirzadi MR, Baderldin S, Abro A, Zaki [164] Şensoy G, Çaltepe Dinler G, Kalkan G, Ateş A, Belet N, Albayrak D. Crimean-
A, Dessi Z, et al. Crimean-Congo hemorrhagic fever in Dubai, United Congo haemorrhagic fever: peritoneal and pleural effusion. Ann Trop Paediatr
Arab Emirates, 2010: case report. Iran Red Crescent Med J 2016;18(8): 2011;31(2):169–72.
e38374. [165] Ture Z, Ulu Kılıç A, Celik I, Tok T, Yağcı-Çağlayık D. Crimean-Congo hemor-
[136] Sheikh AS, Sheikh AA, Sheikh NS, Rafi-U-Shan null, Asif M, Afridi F, et al. rhagic fever with hyperbilirubinemia and ascites: an unusual presentation. J
Bi-annual surge of Crimean-Congo haemorrhagic fever (CCHF): a five-year Med Virol 2016;88(1):159–62.
experience. Int J Infect Dis 2005;9(1):37–42. [166] Gharabaghi MA, Chinikar S, Ghiasi SM, Morady M, Ahmadinejhad T, Paydary
[137] Jamil B, Hasan RS, Sarwari AR, Burton J, Hewson R, Clegg C. Crimean-Congo K. Severe Crimean-Congo haemorrhagic fever presented with massive
hemorrhagic fever: experience at a tertiary care hospital in Karachi, Pakistan. retroperitoneal haemorrhage that recovered without antiviral treatment. BMJ
Trans R Soc Trop Med Hyg 2005;99(8):577–84. Case Rep 2011;19:2011.
[138] Saleem J, Usman M, Nadeem A, Sethi SA, Salman M. Crimean-Congo hem- [167] Aksoy HZ, Yilmaz G, Aksoy F, Koksal I. Crimean-Congo haemor-
orrhagic fever: a first case from Abbottabad, Pakistan. Int J Infect Dis rhagic fever presenting as epididymo-orchitis. J Clin Virol 2010;48(4):
2009;13(3):e121–3. 282–4.
[139] Malik S, Diju IU, Naz F. Crimean Congo hemorrhagic fever in Hazara division. [168] Kaya S, Yilmaz G, Ertunç B, Koksal I. Parotitis associated with Crimean Congo
J Ayub Med Coll Abbottabad 2011;23(2):90–2. hemorrhagic fever virus. J Clin Virol 2012;53(2):159–61.
[140] Ali F, Saleem T, Khalid U, Mehmood SF, Jamil B. Crimean-Congo hemorrhagic [169] Özer S, Kazancı NÖ, Sönmezgöz E, Karaaslan E, Yılmaz R. Syndrome of inap-
fever in a dengue-endemic region: lessons for the future. J Infect Dev Ctries propriate antidiuretic hormone associated with Crimean-Congo hemorrhagic
2010;4(7):459–63. fever. Turk Parazitolojii Derg 2014;38(4):275–7.
P. Fillâtre et al. / Médecine et maladies infectieuses 49 (2019) 574–585 585

[170] Kilicli F, Dokmetas HS, Dokmetas İ. Acute evaluation of pituitary function [172] Deveci K, Uysal EB, Kaya A, Sancakdar E, Alkan F. Evaluation of renal involve-
in patients with Crimean-Congo haemorrhagic fever. Clin Endocrinol (Oxf) ment in children with Crimean-Congo hemorrhagic fever. Jpn J Infect Dis
2012;76(2):241–5. 2013;66(6):493–6.
[171] Kleib AS, Salihy SM, Ghaber SM, Sidiel BW, Sidiya KC, Bettar ES. Crimean- [173] Ahmeti S, Ajazaj-Berisha L, Halili B, Shala A. Acute arthritis in crimean-congo
Congo hemorrhagic fever with acute subdural hematoma, Mauritania, 2012. hemorrhagic fever. J Glob Infect Dis 2014;6(2):79–81.
Emerg Infect Dis 2016;22(7):1305–6.

You might also like