Dermatologic Manifestations of Viral Hemorrhagic Fevers

Back

Background

Viral hemorrhagic fevers (VHFs) are a group of etiologically diverse viral diseases unified by common underlying pathophysiology. These febrile diseases result from infection by viruses from four viral families: Arenaviridae, Bunyaviridae, Filoviridae, and Flaviviridae.[1]

The viruses in the four families are all RNA viruses. All share the feature of having a lipid envelope. Survival and perpetuation of the viruses is dependent on an animal host known as a natural reservoir; humans are not the natural reservoir. With the exception of a vaccine for yellow fever and ribavirin, which is used as a drug treatment for some arenaviral infections, no cures or drug treatments for viral hemorrhagic fever exist. Only supportive treatment is possible.

Not all viruses in these families cause viral hemorrhagic fever. Viral hemorrhagic fevers share certain clinical manifestations, regardless of the virus that causes the disease. However, different viruses can cause a range of various clinical problems in addition to viral hemorrhagic fever. Common clinical manifestations of viral hemorrhagic fever are increased capillary permeability, leukopenia, and thrombocytopenia. Viral hemorrhagic fever is manifested by sudden onset, fever, headache, generalized myalgia, backache, petechiae, conjunctivitis, and severe prostration. Various hemorrhagic symptoms follow, ultimately resulting in focal inflammatory reaction and necrosis with leukocytosis.

Although the viruses are distributed all over the world, they have a higher occurrence in tropical areas, such as South America, Africa, and the Pacific Islands. They have a higher likelihood of importation because of increased travel and scientific research involving the use of imported tropical animals, which often serve as intermediate hosts. The viruses are transmitted by two main categories of natural reservoirs: arthropods and rodents. Arenaviruses and Hantavirus (a Bunyavirus) are primarily rodent-borne, whereas flaviviruses, as well as nairoviruses and phleboviruses (both bunyaviruses), are arthropod-borne.

Transmission occurs mainly by means of contact with the following: natural reservoirs (eg, mosquito bites, rodent bites); reservoir excretions, secretions, or blood; aerosolized particles contaminated by reservoir secretions, excretions, or blood; or intermediate hosts (eg, monkeys, livestock) or their excretions, secretions, or blood. Person-to-person transmission and nosocomial transmission also occur. Nosocomial outbreaks are not uncommon in developing countries, where safe infectious disease practices have not been implemented and supplies are in shortage.

See the Medscape articles CBRNE - Viral Hemorrhagic Fevers and Pediatric Viral Hemorrhagic Fevers for more information.

Also see Ebola: Care, Recommendations, and Protecting Practitioners, a Critical Images slideshow, to review treatment, recommendations, and safeguards for healthcare personnel.

Pathophysiology

Despite the diverse taxonomy of the four virus families involved in viral hemorrhagic fevers (VHFs), they share some common characteristics. They are all RNA viruses that have a lipid envelope, rendering them relatively susceptible to detergents and a low-pH environment, as well as household bleach. On the other hand, they are quite stable at neutral pH; this factor helps these viruses to stay stable in blood for a long period, which allows them to be isolated from a patient’s blood after weeks of storage at refrigerator temperature. In addition, these viruses are stable as fine-particle aerosols, which renders them highly infectious.

Viral-targeted cells in the body include monocytes, dendritic cells, macrophages, and vascular endothelial cells, which then disseminate through lymphatics to other organs.[2]

Recognition of viral infection by the innate immune system occurs through the cytoplasmic recognition of cellular receptors of viral nucleic acids. Following this recognition and activation of cellular receptors, type I interferon is activated, resulting in initiation of interferon signaling.[3]

The main common underlying pathophysiologic feature of viral hemorrhagic fevers is that the vascular bed is attacked, with resultant microvascular damage and changes in vascular permeability. However, specific pathophysiologic findings can vary depending on the virus family and the species involved.

In general, an initial febrile illness is followed by hemorrhaging into the skin and the mucous membranes, hemorrhagic rashes, and hemorrhaging from body orifices, especially gastrointestinal and genitourinary bleeding. Lassa fever, although fatal, is not characterized by significant bleeding. Other clinical findings include thrombocytopenia and leukocytopenia.

Ebola (Filoviridae) viral protein VP35 was found to inhibit interferon regulatory factor 3, which is necessary for the induction of an antiviral immune response and interferon. Ebola virus was also found to alter the immune signaling pathways through its ability to interfere with dendritic cells that link adaptive and innate immune responses,[4] in addition to the release of extensive cytokines, which cause endothelial damage, coagulopathy, and, finally, multiorgan failure.[2]

Similarities regarding the pattern of cytokine production between Ebola virus disease and Crimean-Congo hemorrhagic fever (CCHF) were noted. Positive correlation was noted between viral load and interleukin (IL)‒10 and monocyte chemoattractant protein (MCP)‒1, and negatively correlated with the IL-12/IL-10 ratio.[5]

In severe dengue fever (Flavivirus), marked capillary permeability and coagulopathy are noted as a result of the immune response.[6] HLA-B44 has been found to be protective against dengue hemorrhagic fever.[7] In contrast, diabetic patients were found to be more susceptible to developing severe dengue hemorrhagic fever and dengue shock syndrome.[8] Circulating immune complexes, serum cryoglobulins, and IL-8 were found to be higher by 9-fold and 2.2-fold in dengue hemorrhagic fever and dengue fever, respectively, compared with healthy individuals. Peak levels of circulating immune complexes, IL-8, and cryoglobulins were found to be associated with thrombocytopenia.[9] Serum IgM levels specific for dengue fever virus were found to be significantly higher in dengue fever when compared with dengue hemorrhagic fever cases, while IgG, IgA, and IgE levels were found to be higher in dengue hemorrhagic fever cases. Higher titer of IgG was found to be associated with lower platelet counts.[10]

In Rift Valley fever (Bunyavirus), IL-8, IL-10, and CXCL9 were detected at significantly higher levels when comparing fatal cases to uninfected individuals and infected survivors.[11]

In Hantavirus (Bunyaviridae) infection, endothelial dysfunction is noted, wherein high levels of endothelial glycocalyx degradation is found to correlate with early disease activity, which eventually results in tissue edema, hypotension, and shock.[12]

Etiology

Causes of viral hemorrhagic fevers are the specific RNA viruses mentioned above. Infection by different viruses results in hemorrhagic fever with different complications, symptoms, and severity, as previously discussed.

Viruses are usually transmitted by mosquitoes, ticks, or rodents. Some species of bats may also prove to be virus carriers.[13]

In a study that included 200 ticks collected from south of Iran analyzed by reverse-transcription polymerase chain reaction for the presence of Crimean-Congo hemorrhagic fever (CCHF) virus genome, viral genome was detected in 4.5% of studied ticks, which indicates that ticks in that area are widely infected and necessitates regular control and monitoring of livestock in order to reduce the dispersion of ticks.[14]

An epidemiologic survey of ticks, rodents, and infected individuals with CCHF in Northern region of Xiniang, China isolated a two new CCHF virus strains, which were different from the previously reported strain in China.[15]

Person-to-person transmission may occur through mucous membranes or through contact with body fluids from the infected patient. Transmission can even occur after the death of the infected person, as their skin is heavily infected.[16]

Contaminated syringes and needles played a role in the recent outbreak of Ebola hemorrhagic fever.

Persistence of Ebola virus in semen was reported in the Ebola virus outbreak of 2014-2015 in three men. The semen was positive for Ebola virus RNA on days 199, 140, and 284 after symptom onset.[17, 18, 19] Evidence of persisting Ebola virus in feces, sweat, saliva, and urine were also reported up to 26 days after initial symptoms.[20, 21]

The strong immunologic response to the viruses may be central to the pathophysiology of plasma leakage associated with these diseases.[22]

Epidemiology

Frequency

United States

Most of the natural reservoirs of these viruses live in tropical areas. Hence, the virus does not typically infect persons in the United States. Random cases of infection occur as a result of the importation of viruses by travelers or the importation of scientific research animal subjects.

Several cases of infection resulting in Hantavirus pulmonary syndrome (HPS), however, have been reported across the United States.[23]

During the 2014 Ebola outbreak, several US healthcare personnel have been infected while in Africa and have been transported to the United States for treatment. A traveller from Liberia also became ill and sought treatment during a visit to Texas, and he later died of the infection. One of his treating nurses then presented with a low-grade fever and tested positive for Ebola virus infection. Further, individuals in several US states who recently travelled to West Africa have developed fever and other symptoms, prompting evaluation for Ebola virus infection at US hospitals.[24]

International

The geographic distribution is dependent on the reservoir host and ecology of each disease.[25]

Table 1. Geographic Distribution of Viral Hemorrhagic Fevers



View Table

See Table

In West Africa, the 2014-2015 Ebola epidemic was first reported in Guinea in March 2014. It was the largest Ebola virus epidemic documented, with a total of 28,220 reported cases and 11,291 deaths (WHO September 16, 2015). In July 2014, a local outbreak was declared in Sierra Leone, and the affected district was the first to be declared Ebola-free by local authorities on January 10, 2015.[26, 27, 28]

Guinea demonstrated consistent low transmissibility and, accordingly, the smallest number of reported cases. Liberia showed the highest level of transmission before October 2014 and remained low since that time. Sierra Leone showed detectable waves of the disease up to mid March 2015, resulting in the largest number of cases reported.[29]

Four confirmed CCHF outbreaks within the past 2 years have been reported in Uganda after more than 50 years of no reported cases of human CCHF.[30]

A Chikungunya outbreak in southeastern Senegal in 2010 included 45 confirmed cases. In addition, 83% of monkeys that were randomly sampled were found to be seropositive, and Chikungunya virus was detected in 42 pools of mosquitos, mainly Aedes furcifer.[31]

A study conducted on 3,322 confirmed dengue cases in Kaohsiung city in Taiwan found that the outbreak of dengue fever was initiated by imported cases from other endemic countries. It took a median of 5 days after the appearance of symptoms for patients to report to a medical facility.[32] A positive correlation was found between confirmed cases and weather parameters (temperature, relative humidity, and rainfall) at a time lag of 1 month and 2 months. This may help in developing an early warning surveillance system.

Poverty was associated with high rates of transmission in a study done in Montserrado County, Liberia.[33]

In an attempt to understand the heterogenicity in dengue disease transmission, a study analyzing 18 years of monthly dengue surveillance was conducted in a total of 273 provinces in eight countries in Southeast Asia. A strong pattern of synchronous transmission across the entire region was detected. This synchrony in dengue incidence was noted to coincide with elevated temperatures in 1997-1998 and the strongest El Niño episode of the century. A low incidence was noted during the period of 2001-2002. Localized travelling waves of epidemic cycles were detected in Laos, Thailand, and the Philippines.[34]

Race

No race is known to be more vulnerable than another to RNA viral infection. Geography is a determining factor.

Sex

Neither sex is known to be more or less vulnerable to RNA viral infection.

Age

Age plays a role in increasing the vulnerability to infection in only 2 circumstances. First, young and elderly persons are more susceptible because of their weaker immune systems. Second, adults are more susceptible if they work in settings in which the exposure risk is increased (eg, clinics or hospitals, agrarian settings).

A shift in dengue fever towards older adults has been noted in the past decade.[35]

Prognosis

Survival may be possible with appropriate support care, depending on the virus.

Table 2. Viral Hemorrhagic Fever Mortality Rates



View Table

See Table

The estimated case fatality rate for the recent Ebola outbreak was 76.4%. The proportion of total deaths in Guinea, Sierra Leone, and Liberia was 21.6%, 35.8%, and 42.5%, respectively. The highest risk of dying was among healthcare workers in areas with intense transmission and countries with insufficient bed capacities. Other factors that enhanced the spread and magnitude of this outbreak were the insufficient enforcement of public health regulations and deplorable healthcare delivery infrastructure in war-ravaged regions.[36]  In the recent Ebola virus disease outbreak in Sierra Leone, it was found that chest pain, symptoms of confusion, coma and viral load greater than 106 copies/mL were significantly associated with a poor prognosis. Viral load was the most important factor that affected the survival of patients from the disease.[37]

A total of 278 human cases were confirmed with Rift Valley fever in the recent outbreak in South Africa in 2010-2011, with 25 deaths.

Children can develop dengue hemorrhagic shock syndrome (DHSS), a complication with a mortality rate of 4-12%.

Patient Education

Educate travelers and geographically vulnerable groups, especially health care workers, agrarian workers, and rural populations, about the following risks:

Educate health care workers and others about the detrimental effects of nosocomial transmission and about how such spread can be prevented by implementing infectious disease safety and contact precautions, such as the following:

Educate health care workers and others about decontamination procedures, such as the use of hypochlorite or phenolic disinfectants.

For patient education resources, see the Bites and Stings Center, as well as Ticks.

History

The nature of each disease depends on the viral strain characteristics and virulence, dose, routes of exposure, and host factors. For example, dengue fever exhibits a hemorrhagic nature only in patients previously exposed to a heterologous dengue serotype.

Not all individuals who are infected have viral hemorrhagic fever. The target organ is the vascular bed. As a result, the dominant clinical manifestations are due to microvascular damage and changes in vascular permeability.

Viral hemorrhagic fever should be suspected in patients who live in or who have returned from locations of viral occurrence if they have severe fever and evidence of vascular involvement (eg, subnormal blood pressure, postural hypotension, petechiae, easy bleeding, flushing, nondependent edema).

The severity of symptoms varies. Common symptoms include the following:

Severe viral hemorrhagic fever evolves to shock and generalized mucous membrane hemorrhage. Viral hemorrhagic fever is often accompanied by neurologic, hematopoietic, or pulmonary involvement. Hepatic damage is also common among viral hemorrhagic fevers, but hepatic failure, as indicated by jaundice and other evidence, is seen in some cases of Rift Valley fever (RVF), Congo-Crimean hemorrhagic fever (CCHF), Marburg fever, Ebola fever, and yellow fever. Neurologic symptoms and thrombocytopenia are common in Argentinian and Bolivian hemorrhagic fevers. Kyasanur Forest disease and Omsk fever are known for their pulmonary involvement and a biphasic illness with central nervous system (CNS) manifestations. Hantavirus pulmonary syndrome (HPS) is known for its pulmonary involvement leading to noncardiogenic pulmonary edema.[23]

Hemorrhagic fevers with renal syndrome may be associated with concomitant central diabetes insipidus. This should be suspected with persistent diuretic phase but normal pituitary MRI.[38]

Physical Examination

In general the illness demonstrates three phases, generalization phase (week 1), early organ phase (week 2), and late organ phase (week 3).

The generalization phase includes fever, chills, sore throat, and gastrointestinal symptoms.

In the early organ phase, endothelial damage starts to occur and is reflected clinically in the form of petechiae, ecchymoses, mucosal hemorrhage, bloody diarrhea, hematemesis, conjunctival congestion, and dyspnea due to pulmonary edema.

The late organ phase shows progression to disseminated intravascular coagulation, shock, and liver and renal failure, which eventually may progress to death.

Arenaviruses [39]

See the image below.



View Image

New World Arenavirus - Machupo. Oral mucosal hyperemia and hemorrhage in a patient with Bolivian hemorrhagic fever.

Lassa, Argentinian, and Bolivian viral hemorrhagic fever

The first signs of Lassa, Argentinian, and Bolivian viral hemorrhagic fever are nonspecific and include fever, headache, and sore throat. The second stage is characterized by an exacerbation of symptoms and rashes on the face or the neck. Gastrointestinal and urogenital tract bleeding and shock may follow during the second week. Neurologic sequelae are a main feature, especially in Lassa hemorrhagic fever.

Argentinian viral hemorrhagic fever

Early Argentinian viral hemorrhagic fever findings include high fever, malaise, headache, retro-orbital pain, anorexia, and nausea. Signs are erythematous rash of the face, the neck, and the thorax, and hemorrhages into the skin and the mucous membranes. CNS involvement appears around day 7 in fatal cases. It is an important indicator of severity and is a better indicator than hemorrhage.

Bolivian viral hemorrhagic fever

Bolivian hemorrhagic fever has an insidious onset, leading to high temperature, severe myalgia, headache, vomiting, retrobulbar pain, conjunctival hyperemia, leukopenia, and mild thrombocytopenia. Petechiae of the skin and the oral mucous membranes usually appear around day 4. Epistaxis, gingival bleeding, hematemesis, melena, and metrorrhagia may follow. Hypotension and/or neurologic abnormalities, including tremors, delirium, and seizures, develop between days 6 and 10. Circulatory collapse eventually results in death.

Lassa fever

Lassa fever can vary from a long-lasting acute fever to a fatal disease. Its clinical manifestations include headache, fever, weakness, myalgia, ulcerative pharyngitis, dysphagia, anorexia, nausea, vomiting, cough, and constipation. The aforementioned findings can be followed by diarrhea, melena, dysuria, cutaneous petechiae and ecchymoses, erythema of the face and the thorax, facial and cervical edema, rales, rhonchi, stridor, wheezing, hypotension, hepatic tenderness, pleural effusions, cloudy sensorium, and seizures. Deafness is a frequent sequela of severe cases

Bunyaviruses

See the image below.



View Image

Bunyavirus infection. Ecchymoses encompassing left upper extremity one week after onset of CCHF. Ecchymoses often are accompanied by hemorrhage in oth....

Rift Valley fever [40]

RVF is an influenzalike disease. Its onset involves chills, fever, headache, retro-orbital pain, myalgia, anorexia, vomiting, and diarrhea. Physical signs include fever, facial erythema, and conjunctival suffusion. Severe RVF can present with the clinical syndrome of fever, large joint arthralgia, and gastrointestinal complaints and is later followed by jaundice, right upper quadrant pain, and delirium.[41] A small percentage of patients present with retinal lesions, which may result in permanent blindness, hemorrhagic diathesis, and encephalitis. RVF may progress to a hemorrhagic fever stage. Clinical manifestations of this stage may include hematemesis, melena, petechial and ecchymotic cutaneous lesions, gingival bleeding, hematuria, epistaxis, and jaundice.

Congo-Crimean hemorrhagic fever

Early symptoms of CCHF include those previously mentioned for RVF. Patients may present with erythema of the pharynx, the conjunctivae, the face, and the neck; a petechial rash on the trunk and the limbs; and hemorrhages of the uvula and the soft palate.

CCHF patients commonly have severe headaches that may resemble migraine. The severity depends on vascular endothelial damage, abnormal release of inflammatory cytokines, and vasodilatation.[42]

CCHF is a severe disease. On days 3-7, patients may have hematemesis; melena; cutaneous purpura; epistaxis; hematuria; hemoptysis; and bleeding from gingival, oral, uterine, and venipuncture sites. Profound disseminated intravascular coagulation is frequently noted

Korean hemorrhagic fever [43]

This infection is a subgroup of hemorrhagic fever with renal syndrome (HFRS). Disease progression is severe and is characterized by fever, hemorrhage, and renal failure. Oliguria has been reported.

Hantavirus pulmonary syndrome

The illness has 5 distinct phases and is marked by deterioration of breathing during the cardiopulmonary phase. Capillary leakage results in noncardiogenic pulmonary edema and may progress to shock. As in HFRS, disease progression is rapid with a high rate of mortality.

Filoviruses

See the image below.



View Image

Filovirus disease - Ebola fever. Patient with Ebola hemorrhagic fever during 1976 outbreak in Zaire demonstrating palatal petechiae and hemorrhage. Ph....

Ebola and Marburg viruses are pantropic, that is, they infect and cause lesions in many organs, especially the liver and the spleen.[44] Both organs become enlarged and dark while undergoing severe degeneration. Disseminated intravascular coagulation may be a feature of late disease. Damage to endothelial cells increases vascular permeability, followed by hemorrhage and shock; these are central features of the infections, which have high mortality rates.

Ebola fever [45, 46]

Early symptoms of Ebola-induced viral hemorrhagic fever are high temperature, headache, myalgia, stomach pain, rashes, sore throat, and red and itchy eyes. Severe watery diarrhea is reported. Rash usually develops at the end of the first week of illness. It is characterized by a morbilliform rash involving the face, neck, arms, and trunk. It may desquamate. This sign was reported rarely in the recent outbreak of Ebola in 2014-2015 in Sierra Leone.[47]

Hemorrhage noted on patients is in the form of petechiae, ecchymoses, blood in the stool, oozing from venipuncture sites, and mucosal hemorrhaging. Major bleeding usually occurs at the terminal stage of the disease. Bleeding was observed in about 20% of patients in the 2014-2015 outbreak.[47]

Within 1 week of disease onset, chest pain, hemorrhaging from body orifices, blindness, and death occur.

In the recent Ebola outbreak in West Africa, the clinical presentation of patients admitted in Connaught Hospital, Sierra Leone, were analyzed in a retrospective cohort study from May 2014 to December 2014.[48] Fever was present in 83% of cases, intense fatigue in 68% of cases, nausea or vomiting in 50% of cases, and diarrhea in 41% of cases. Of confirmed Ebola virus disease cases, 9% had no history of fever and denied exposure to any risk factor for Ebola virus disease.

Marburg fever [49]

Disease onset is marked by fever, chills, headache, and myalgia. Approximately 5 days after disease onset, a maculopapular rash may appear, with greatest prominence on the trunk. Patients present with jaundice, inflammation of the pancreas, weight loss, delirium, shock, liver failure, and multiorgan dysfunction.

Flaviviruses

See the image below.



View Image

Patient with morbilliform exanthem of dengue fever. Note islands of sparing characteristics for dengue. Photo courtesy Duane Gubler, PhD.

Omsk hemorrhagic fever

Physical findings include congested conjunctivae, papulovesicular eruptions on the soft palate (an important diagnostic sign), severe pain in the back and the limbs, fever, headache, diarrhea, and vomiting. The illness is biphasic, with meningismus or meningoencephalitis occurring in the second phase. The disease is also characterized by hyperemia of the face and the upper body. Bleeding gums, epistaxis, hemoptysis, uterine hemorrhage, hematemesis, melena, and other hemorrhaging may occur.

Yellow fever

Yellow fever is characterized by jaundice, a result of midzone necrosis of the liver (by about the third day), flushing of the face, congested conjunctivae, and reddening edges of the tongue. The heart and the kidney are damaged. Hemorrhagic symptoms may appear early, with swelling and bleeding of the gums and epistaxis. Hemorrhaging from the gastrointestinal mucosa may cause black vomit and melena.

Dengue fever [50, 51, 52]

Patients present with maculopapular rashes and fever, severe joint and muscle pains, lymphadenopathy, and altered or diminishing gustation.

The fever is characteristically biphasic, breaking and returning in 1-2 days.[53] Dengue fever, like yellow fever, may progress from a febrile illness to a hemorrhagic one in about 5% of cases. Minor hemorrhagic symptoms, such as epistaxis, petechiae, or bleeding gums, may accompany other febrile symptoms early in the disease. An enlarged liver can be palpated during the later stages of the disease. Signs of worsening of Dengue fever include ongoing vomiting, worsening abdominal pain, mucosal bleeding, enlargement of the liver, lethargy or restlessness, serosal effusion, and high hematocrit combined with low platelet count.[53] Children may develop DHSS, a complication with a mortality rate of 4-12%. Transient left ventricular systolic and diastolic dysfunction were found to be common in hospitalized children with dengue fever and was related to the severity of plasma leakage.[54] However, cardiac structural changes were not common.

In a retrospective study that included 667 dengue patients, fever, myalgia, headache, arthralgia, abdominal pain, vomiting, and rash were observed in more than 40% of patients.[55] Dehydration, rash, pleural effusion, shortness of breath, and thick gall bladder were found to be significantly associated with dengue hemorrhagic fever when compared with dengue fever. Predictors noted for dengue hemorrhagic fever included older age, diabetes mellitus, secondary infection, thick gallbladder, lethargy, and delayed hospitalization.

Complications

Rift Valley hemorrhagic fever may lead to blindness in some cases. Individuals who are infected can occasionally have encephalitis due to bunyavirus or flavivirus infections. By the time the patient presents with encephalitis, serum antibody levels are usually detectable. Lassa and Machupo viruses can cause nerve deafness. Patients may develop bacterial sepsis or respiratory failure from fluid resuscitation. Multisystem shock leading to death is possible.

In a study analyzing 277 survivors of the Ebola epidemic in Sierra Leone, clinical sequelae were noted and include arthralgia in 76% of patients, new ocular symptoms in 60% of cases, uveitis in 18% of patients, and auditory symptoms in 24% of cases.[56] A higher viral load at time of presentation was significantly associated with uveitis and with the development of new ocular symptoms. Late-onset encephalitis, alopecia, paraesthesia, depression or anxiety, and polyarthritis were also reported.[57, 58]

Severe rhabdomyolysis, acute kidney injury, immune thrombocytopenia, and postencephalitic parkinsonism are complications reported following dengue virus infection.[59, 60, 61, 62]

In Crimean-Congo hemorrhagic fever (CCHF), cardiac hypokinesia, pericardial effusion, T-wave changes, myocardial involvement, and bundle-branch block are complications reported.[63]

Laboratory Studies

Although the clinical findings can suggest a hemorrhagic fever, laboratory studies are required to identify disease, to distinguish it from other conditions, and to confirm its etiology.

As a rule, clinical blood and/or urine tests reveal leukopenia (except in Lassa fever, Hantaan viral fever, and some severe cases of CCHF), thrombocytopenia (except in Lassa fever), and proteinuria and/or hematuria (in Argentinian viral hemorrhagic fever, Bolivian viral hemorrhagic fever, and hemorrhagic fever with renal syndrome [HFRS]; common in other viral hemorrhagic fevers). Such tests include the following:

Specialized infectious disease containment is required for the safe handling of these viruses. Biochemical tests are available for the rapid detection of viral antigen during viremia or in postmortem specimens. Such tests include the following:

Other Tests

Electron microscopy of the infected tissue may be helpful.

Medical Care

Supportive care

Rapidly transfer the patient to a hospital with minimal trauma in order to prevent trauma to the fragile capillary bed.

The judicious use of sedative, pain-relieving, and amnesic medications can be helpful in managing malaise, confusion, myalgia, and hyperesthesia. Intramuscular injections and the use of aspirin and other anticoagulant drugs should be avoided.

Adequate hydration reduces the death rate, and oral rehydration may not be adequate.

Intensive supportive care is necessary for most cases of viral hemorrhagic fever. General supportive care principles apply to the treatment of hemodynamic, hematologic, pulmonary, and neurologic manifestations of viral hemorrhagic fever. Supportive care entails maintaining the patient's oxygen status and blood pressure and balancing fluid and electrolyte levels. Blood, platelet, and plasma replacement and management can be crucial, depending on the case. Convalescent plasma infusions may be effective in Argentinian hemorrhagic fever, if they are administered within the first 8 days of onset.

In Dengue fever, for which the main pathology is capillary leakage, brisk infusion with crystalloids should be instituted, followed by colloids or albumin if there is no response.

In Hantavirus infection, acute renal failure recovery may be accompanied by severe polyuria that should be managed carefully with close observation of fluid balance and electrolytes values.

Antiviral therapy with ribavirin may be useful in several viral hemorrhagic fevers, especially those caused by Arenaviruses. Although ribavirin inhibits viral DNA and RNA synthesis, it is not sensitive to the replication mechanisms of all RNA viruses. Ribavirin is proven effective in the treatment of Lassa fever and Congo-Crimean hemorrhagic fever.[66] It is somewhat effective in the treatment of other arenavirus and Hantavirus infections, in which it decreases mortality rates when used early in the course of the disease. Additionally, discovery of compounds with antiflaviviral activity have shown promise. Currently, several ribavirin analogues are undergoing clinical trials.[67]

Interferon-alfa has shown promising results in Arenaviral infections in vitro as an adjunctive therapy with ribavirin.[68]

Transfer

Patients may require treatment for secondary infections that may arise. Intensive care management may be required for viral hemorrhagic fevers.[69]

If hospitals or clinics are not equipped to deal with such infectious diseases, patients should be transferred to facilities with the following:

Patient transfer in infectious disease cases may increase the chances of nosocomial transmission if proper precautions are not taken.

Vaccination

Vaccination may be considered. The only approved vaccine is for yellow fever. The WHO Strategic Advisory Group recommends a single primary dose for most travelers; a booster dose of the vaccine is no longer required, as the vaccine was found to confer sustained immunity and lifelong protection against yellow fever disease.[70]

Nonapproved vaccines include that developed for Argentinian viral hemorrhagic fever. This vaccine is a live-attenuated, investigational vaccine. It also seems to offer protection against Bolivian viral hemorrhagic fever. Both inactivated and live-attenuated vaccines for RVF are under investigation.[71] No vaccines are currently available for filovirus infection or dengue. Preliminary results suggest potential for successful prevaccination and postvaccination exposure against Ebola and Marburg viruses.[13, 72, 73]

Caregiver exposure

Persons percutaneously or mucocutaneously exposed to blood, excretions, or secretions of individuals who are infected should wash the affected areas with soap and water. Affected mucous membranes should be irrigated with water or sodium chloride solution.

Further reading

Additional information is available from the Centers for Disease Control and Prevention. See Infection Control for Viral Haemorrhagic Fevers in the African Health Care Setting and Questions and Answers on Ebola.

Consultations

Consultation with the following specialists may be warranted:

Diet

Fluid and electrolyte balance should be maintained.

Activity

Malaise and myalgia, among other symptoms, dictate bed rest restrictions.

Prevention

Prevention involves the following:

Deterrence involves the eradication of rodent and arthropod vectors.

Medication Summary

The goals of pharmacotherapy are to induce remission, to reduce morbidity, and to prevent complications.

Ribavirin (Rebetol)

Clinical Context:  Ribavirin is suggested drug treatment for Hantavirus infections and Arenavirus infections, especially Lassa fever and Crimean-Congo hemorrhagic fever. It is especially effective when administered in the early course of the disease. It inhibits viral replication by inhibiting DNA and RNA synthesis.

Class Summary

Antiviral agents inhibit the replication of RNA and DNA viruses.

Author

Amira M Elbendary, MBBCh, MSc, Visiting Dermatopathology Fellow, Ackerman Academy of Dermatopathology; Assistant Lecturer, Department of Dermatology, Kasr Alainy University Hospitals, Cairo University, Egypt

Disclosure: Nothing to disclose.

Coauthor(s)

Dirk M Elston, MD, Professor and Chairman, Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina College of Medicine

Disclosure: Nothing to disclose.

Specialty Editors

David F Butler, MD, Former Section Chief of Dermatology, Central Texas Veterans Healthcare System; Professor of Dermatology, Texas A&M University College of Medicine; Founding Chair, Department of Dermatology, Scott and White Clinic

Disclosure: Nothing to disclose.

Jeffrey P Callen, MD, Professor of Medicine (Dermatology), Chief, Division of Dermatology, University of Louisville School of Medicine

Disclosure: Received honoraria from UpToDate for author/editor; Received royalty from Elsevier for book author/editor; Received dividends from trust accounts, but I do not control these accounts, and have directed our managers to divest pharmaceutical stocks as is fiscally prudent from Stock holdings in various trust accounts include some pharmaceutical companies and device makers for i inherited these trust accounts; for: Allergen; Celgene; Pfizer; 3M; Johnson and Johnson; Merck; Abbott Laboratories; AbbVie; Procter and Gamble; Amgen.

Chief Editor

William D James, MD, Paul R Gross Professor of Dermatology, Vice-Chairman, Residency Program Director, Department of Dermatology, University of Pennsylvania School of Medicine

Disclosure: Received income in an amount equal to or greater than $250 from: Elsevier; WebMD.

Acknowledgements

Ally N Alai, MD, FAAD Former Professor, Department of Dermatology, Clinical Faculty and Preceptor, Department of Family Practice, University of California, Irvine, School of Medicine; Former Professor and Preceptor, Department of Family Practice Residency Training, Downey Medical Center; Medical Director, The Skin Center at Laguna; Expert Medical Reviewer, Medical Board of California; Expert Consultant, California Department of Consumer Affairs; Expert Reviewer, California Department of Registered Nursing

Ally N Alai, MD, FAAD is a member of the following medical societies: American Academy of Dermatology and American Society for MOHS Surgery

Disclosure: Nothing to disclose.

Eiman N Anvari, DO, MSc Resident Physician, Department of Radiology, Philadelphia College of Osteopathic Medicine

Eiman N Anvari, DO, MSc is a member of the following medical societies: American College of Physicians, American Medical Student Association/Foundation, and American Osteopathic Association

Disclosure: Nothing to disclose.

Arash M Saemi, MD Resident Physician, Department of Radiology, Dartmouth-Hitchcock Medical Center

Arash M Saemi, MD is a member of the following medical societies: American College of Physicians, Radiological Society of North America, Sigma Xi, and Society of Interventional Radiology

Disclosure: Nothing to disclose.

References

  1. Racsa LD, Kraft CS, Olinger GG, Hensley LE. Viral Hemorrhagic Fever Diagnostics. Clin Infect Dis. 2016 Jan 15. 62 (2):214-9. [View Abstract]
  2. Feldmann H, Geisbert TW. Ebola haemorrhagic fever. Lancet. 2011 Mar 5. 377(9768):849-62. [View Abstract]
  3. Randall RE, Goodbourn S. Interferons and viruses: an interplay between induction, signalling, antiviral responses and virus countermeasures. J Gen Virol. 2008 Jan. 89 (Pt 1):1-47. [View Abstract]
  4. Kumar A. Ebola virus altered innate and adaptive immune response signalling pathways: Implications for novel therapeutic approaches. Infect Disord Drug Targets. 2016 Jan 7. [View Abstract]
  5. Papa A, Tsergouli K, Çağlayık DY, Bino S, Como N, Uyar Y, et al. Cytokines as biomarkers of Crimean-Congo hemorrhagic fever. J Med Virol. 2016 Jan. 88 (1):21-7. [View Abstract]
  6. Simmons CP, Farrar JJ, Nguyen vV, Wills B. Dengue. N Engl J Med. 2012 Apr 12. 366(15):1423-32. [View Abstract]
  7. Vejbaesya S, Thongpradit R, Kalayanarooj S, Luangtrakool K, Luangtrakool P, Gibbons RV, et al. HLA Class I Supertype Associations With Clinical Outcome of Secondary Dengue Virus Infections in Ethnic Thais. J Infect Dis. 2015 Mar 4. [View Abstract]
  8. Chen CY, Lee MY, Lin KD, Hsu WH, Lee YJ, Hsiao PJ, et al. Diabetes mellitus increases severity of thrombocytopenia in dengue-infected patients. Int J Mol Sci. 2015 Feb 10. 16(2):3820-30. [View Abstract]
  9. Patra G, Ghosh M, Modak D, Bandopadhyay B, Saha B, Mukhopadhyay S. Status of circulating immune complexes, IL8 titers and cryoglobulins in patients with dengue infection. Indian J Exp Biol. 2015 Nov. 53 (11):719-25. [View Abstract]
  10. Bachal R, Alagarasu K, Singh A, Salunke A, Shah P, Cecilia D. Higher levels of dengue-virus-specific IgG and IgA during pre-defervescence associated with primary dengue hemorrhagic fever. Arch Virol. 2015 Oct. 160 (10):2435-43. [View Abstract]
  11. Jansen van Vuren P, Shalekoff S, Grobbelaar AA, Archer BN, Thomas J, Tiemessen CT, et al. Serum levels of inflammatory cytokines in Rift Valley fever patients are indicative of severe disease. Virol J. 2015 Oct 6. 12:159. [View Abstract]
  12. Connolly-Andersen AM, Thunberg T, Ahlm C. Endothelial activation and repair during hantavirus infection: association with disease outcome. Open Forum Infect Dis. 2014 Mar. 1(1):ofu027. [View Abstract]
  13. Bausch DG, Sprecher AG, Jeffs B, Boumandouki P. Treatment of Marburg and Ebola hemorrhagic fevers: a strategy for testing new drugs and vaccines under outbreak conditions. Antiviral Res. 2008 Apr. 78(1):150-61. [View Abstract]
  14. Farhadpour F, Telmadarraiy Z, Chinikar S, Fakoorziba MR, Akbarzadeh K, Moemenbellah-Fard MD, et al. Molecular Detection of Crimean-Congo Hemorrhagic Fever Virus in Ticks Collected From Infested Livestock Populations in a New Endemic Area, South of Iran. Trop Med Int Health. 2016 Jan 13. [View Abstract]
  15. Zhang Y, Shen S, Fang Y, Liu J, Su Z, Liang J, et al. Isolation, Characterization, and Phylogenetic Analysis of Two New Crimean-Congo Hemorrhagic Fever Virus Strains from the Northern Region of Xinjiang Province, China. Virol Sin. 2018 Feb. 33 (1):74-86. [View Abstract]
  16. World Health Organization. Ebola haemorrhagic fever in Zaire, 1976. Bull World Health Organ. 1978. 56(2):271-93. [View Abstract]
  17. Christie A, Davies-Wayne GJ, Cordier-Lassalle T, et al. Possible sexual transmission of Ebola virus - Liberia, 2015. MMWR Morb Mortal Wkly Rep. 2015 May 8. 64 (17):479-81. [View Abstract]
  18. Fink S. Ebola Researchers Take New Look at Risk of Sexual Transmission. NYTimes.com. Available at http://www.nytimes.com/2015/04/17/world/africa/ebola-researchers-take-new-look-at-risk-of-sexual-transmission.html. April 16, 2015; Accessed: February 3, 2015.
  19. Deen GF, Knust B, Broutet N, et al. Ebola RNA Persistence in Semen of Ebola Virus Disease Survivors - Preliminary Report. N Engl J Med. 2015 Oct 14. [View Abstract]
  20. Formenty P, Leroy EM, Epelboin A, Libama F, Lenzi M, Sudeck H, et al. Detection of Ebola virus in oral fluid specimens during outbreaks of Ebola virus hemorrhagic fever in the Republic of Congo. Clin Infect Dis. 2006 Jun 1. 42 (11):1521-6. [View Abstract]
  21. Dowell SF, Mukunu R, Ksiazek TG, Khan AS, Rollin PE, Peters CJ. Transmission of Ebola hemorrhagic fever: a study of risk factors in family members, Kikwit, Democratic Republic of the Congo, 1995. Commission de Lutte contre les Epidémies à Kikwit. J Infect Dis. 1999 Feb. 179 Suppl 1:S87-91. [View Abstract]
  22. Noisakran S, Perng GC. Alternate hypothesis on the pathogenesis of dengue hemorrhagic fever (DHF)/dengue shock syndrome (DSS) in dengue virus infection. Exp Biol Med (Maywood). 2008 Apr. 233(4):401-8. [View Abstract]
  23. Jonsson CB, Hooper J, Mertz G. Treatment of hantavirus pulmonary syndrome. Antiviral Res. 2008 Apr. 78(1):162-9. [View Abstract]
  24. Centers for Disease Control and Prevention. Review of Human-to-Human Transmission of Ebola Virus. Available at http://www.cdc.gov/vhf/ebola/transmission/human-transmission.html. Accessed: October 17, 2014.
  25. Fhogartaigh CN, Aarons E. Viral haemorrhagic fever. Clin Med. 2015 Feb. 15(1):61-6. [View Abstract]
  26. Baize S, Pannetier D, Oestereich L, et al. Emergence of Zaire Ebola virus disease in Guinea. N Engl J Med. 2014 Oct 9. 371 (15):1418-25. [View Abstract]
  27. Dixon MG, Schafer IJ, Centers for Disease Control and Prevention (CDC). Ebola viral disease outbreak--West Africa, 2014. MMWR Morb Mortal Wkly Rep. 2014 Jun 27. 63 (25):548-51. [View Abstract]
  28. WHO Ebola Response Team. Ebola virus disease in West Africa--the first 9 months of the epidemic and forward projections. N Engl J Med. 2014 Oct 16. 371 (16):1481-95. [View Abstract]
  29. Hsieh YH. Temporal Course of 2014 Ebola Virus Disease (EVD) Outbreak in West Africa Elucidated through Morbidity and Mortality Data: A Tale of Three Countries. PLoS One. 2015. 10 (11):e0140810. [View Abstract]
  30. Balinandi S, Patel K, Ojwang J, Kyondo J, Mulei S, Tumusiime A, et al. Investigation of an isolated case of human Crimean-Congo hemorrhagic fever in Central Uganda, 2015. Int J Infect Dis. 2018 Mar. 68:88-93. [View Abstract]
  31. Sow A, Faye O, Diallo M, Diallo D, Chen R, Faye O, et al. Chikungunya Outbreak in Kedougou, Southeastern Senegal in 2009-2010. Open Forum Infect Dis. 2018 Jan. 5 (1):ofx259. [View Abstract]
  32. Chang CJ, Chen CS, Tien CJ, Lu MR. Epidemiological, clinical and climatic characteristics of dengue fever in Kaohsiung City, Taiwan with implication for prevention and control. PLoS One. 2018. 13 (1):e0190637. [View Abstract]
  33. Fallah MP, Skrip LA, Gertler S, Yamin D, Galvani AP. Quantifying Poverty as a Driver of Ebola Transmission. PLoS Negl Trop Dis. 2015 Dec. 9 (12):e0004260. [View Abstract]
  34. van Panhuis WG, Choisy M, Xiong X, et al. Region-wide synchrony and traveling waves of dengue across eight countries in Southeast Asia. Proc Natl Acad Sci U S A. 2015 Oct 20. 112 (42):13069-74. [View Abstract]
  35. Tiawilai T, Tiawilai A, Nunthanid S. Dengue Infection in Elderly Patients. Southeast Asian J Trop Med Public Health. 2015. 46 Suppl 1:165-8. [View Abstract]
  36. Fasina FO, Adenubi OT, Ogundare ST, Shittu A, Bwala DG, Fasina MM. Descriptive analyses and risk of death due to Ebola Virus Disease, West Africa, 2014. J Infect Dev Ctries. 2015 Dec 30. 9 (12):1298-307. [View Abstract]
  37. Zhang X, Rong Y, Sun L, Liu L, Su H, Zhang J, et al. Prognostic Analysis of Patients with Ebola Virus Disease. PLoS Negl Trop Dis. 2015 Sep. 9 (9):e0004113. [View Abstract]
  38. Ahn HJ, Chung JH, Kim DM, Yoon NR, Kim CM. Hemorrhagic fever with renal syndrome accompanied by panhypopituitarism and central diabetes insipidus: a case report. J Neurovirol. 2018 Mar 5. [View Abstract]
  39. Southern PJ. Arenaviridae: the viruses and their replication. Fields BN, Knipe DN, Howley PM, et al, eds. Fields Virology. 3rd ed. Philadelphia, Pa: Lippincott-Raven; 1996. 1505-19.
  40. Peters CJ, Linthicum KJ. Rift valley fever. Handbook of Zoonoses. Boca Raton, Fla: CRC Press; 1994. 129-43.
  41. Kahlon SS, Peters CJ, Leduc J, Muchiri EM, Muiruri S, Njenga MK. Severe Rift Valley fever may present with a characteristic clinical syndrome. Am J Trop Med Hyg. 2010 Mar. 82(3):371-5. [View Abstract]
  42. Aksoy D, Barut H, Duygu F, Çevik B, Kurt S, Sümbül O. Characteristics of headache and its relationship with disease severity in patients with Crimean-Congo hemorrhagic fever. Agri. 2018 Jan. 30 (1):12-17. [View Abstract]
  43. Seo JH, Park KH, Lim JY, Youn HS. Hemorrhagic fever with renal syndrome (HFRS, Korean hemorrhagic fever). Pediatr Nephrol. 2007 Jan. 22(1):156-7. [View Abstract]
  44. Emanuel J, Marzi A, Feldmann H. Filoviruses: Ecology, Molecular Biology, and Evolution. Adv Virus Res. 2018. 100:189-221. [View Abstract]
  45. Chowell G, Nishiura H. Transmission dynamics and control of Ebola virus disease (EVD): a review. BMC Med. 2014 Oct 10. 12(1):196. [View Abstract]
  46. Gire SK, Goba A, Andersen KG, Sealfon RS, Park DJ, Kanneh L, et al. Genomic surveillance elucidates Ebola virus origin and transmission during the 2014 outbreak. Science. 2014 Sep 12. 345(6202):1369-72. [View Abstract]
  47. Schieffelin JS, Shaffer JG, Goba A, Gbakie M, Gire SK, Colubri A, et al. Clinical illness and outcomes in patients with Ebola in Sierra Leone. N Engl J Med. 2014 Nov 27. 371(22):2092-100. [View Abstract]
  48. Lado M, Walker NF, Baker P, Haroon S, Brown CS, Youkee D, et al. Clinical features of patients isolated for suspected Ebola virus disease at Connaught Hospital, Freetown, Sierra Leone: a retrospective cohort study. Lancet Infect Dis. 2015 Sep. 15 (9):1024-33. [View Abstract]
  49. Feldmann H. Marburg hemorrhagic fever--the forgotten cousin strikes. N Engl J Med. 2006 Aug 31. 355(9):866-9. [View Abstract]
  50. Green S, Rothman A. Immunopathological mechanisms in dengue and dengue hemorrhagic fever. Curr Opin Infect Dis. 2006 Oct. 19(5):429-36. [View Abstract]
  51. No Authors Listed. Dengue haemorrhagic fever: early recognition, diagnosis and hospital management--an audiovisual guide for health-care workers responding to outbreaks. Wkly Epidemiol Rec. 2006 Sep 22. 81(38):362-3. [View Abstract]
  52. Munir MA, Alam SE, Khan ZU, Saeed Q, Arif A, Iqbal R, et al. Dengue fever in patients admitted in tertiary care hospitals in Pakistan. J Pak Med Assoc. 2014 May. 64(5):553-9. [View Abstract]
  53. Heilman JM, De Wolff J, Beards GM, Basden BJ. Dengue fever: a Wikipedia clinical review. Open Med. 2014. 8(4):e105-15. [View Abstract]
  54. Kirawittaya T, Yoon IK, Wichit S, Green S, Ennis FA, Gibbons RV, et al. Evaluation of Cardiac Involvement in Children with Dengue by Serial Echocardiographic Studies. PLoS Negl Trop Dis. 2015 Jul. 9 (7):e0003943. [View Abstract]
  55. Mallhi TH, Khan AH, Adnan AS, Sarriff A, Khan YH, Jummaat F. Clinico-laboratory spectrum of dengue viral infection and risk factors associated with dengue hemorrhagic fever: a retrospective study. BMC Infect Dis. 2015 Sep 30. 15:399. [View Abstract]
  56. Mattia JG, Vandy MJ, Chang JC, et al. Early clinical sequelae of Ebola virus disease in Sierra Leone: a cross-sectional study. Lancet Infect Dis. 2015 Dec 22. [View Abstract]
  57. Howlett P, Brown C, Helderman T, Brooks T, Lisk D, Deen G, et al. Ebola Virus Disease Complicated by Late-Onset Encephalitis and Polyarthritis, Sierra Leone. Emerg Infect Dis. 2016 Jan. 22 (1):150-2. [View Abstract]
  58. Epstein L, Wong KK, Kallen AJ, Uyeki TM. Post-Ebola Signs and Symptoms in U.S. Survivors. N Engl J Med. 2015 Dec 17. 373 (25):2484-6. [View Abstract]
  59. Siriyakorn N, Insiripong S. Fatal Rhabdomyolysis in Dengue Hemorrhagic Fever: A Case Report. Southeast Asian J Trop Med Public Health. 2015. 46 Suppl 1:149-52. [View Abstract]
  60. Vachvanichsanong P, McNeil E. Electrolyte Disturbance and Kidney Dysfunction in Dengue Viral Infection. Southeast Asian J Trop Med Public Health. 2015. 46 Suppl 1:108-17. [View Abstract]
  61. Ramírez-Fonseca T, Segarra-Torres A, Jaume-Anselmi F, Ramírez-Rivera J. Dengue Fever: A Rare Cause Of Immune Thrombocytopenia. Bol Asoc Med P R. 2015 Apr-Jun. 107 (2):51-3. [View Abstract]
  62. Bopeththa BVKM, Ralapanawa U. Post encephalitic parkinsonism following dengue viral infection. BMC Res Notes. 2017 Nov 29. 10 (1):655. [View Abstract]
  63. Gülhan B, Kanık-Yüksek S, Çetin İİ, Özkaya-Parlakay A, Tezer H. Myocarditis in a Child with Crimean-Congo Hemorrhagic Fever. Vector Borne Zoonotic Dis. 2015 Sep. 15 (9):565-7. [View Abstract]
  64. Kurosaki Y, Grolla A, Fukuma A, Feldmann H, Yasuda J. Development and evaluation of a simple assay for Marburg virus detection using a reverse transcription-loop-mediated isothermal amplification method. J Clin Microbiol. 2010 Jul. 48(7):2330-6. [View Abstract]
  65. Leroy EM, Baize S, Lu CY, McCormick JB, Georges AJ, Georges-Courbot MC, et al. Diagnosis of Ebola haemorrhagic fever by RT-PCR in an epidemic setting. J Med Virol. 2000 Apr. 60 (4):463-7. [View Abstract]
  66. Ergonul O. Treatment of Crimean-Congo hemorrhagic fever. Antiviral Res. 2008 Apr. 78(1):125-31. [View Abstract]
  67. Ghosh D, Basu A. Present perspectives on flaviviral chemotherapy. Drug Discov Today. 2008 Jul. 13(13-14):619-24. [View Abstract]
  68. Morrill JC, Jennings GB, Cosgriff TM, Gibbs PH, Peters CJ. Prevention of Rift Valley fever in rhesus monkeys with interferon-alpha. Rev Infect Dis. 1989 May-Jun. 11 Suppl 4:S815-25. [View Abstract]
  69. Kothari VM, Karnad DR, Bichile LS. Tropical infections in the ICU. J Assoc Physicians India. 2006 Apr. 54:291-8. [View Abstract]
  70. Staples JE, Bocchini JA Jr, Rubin L, Fischer M, Centers for Disease Control and Prevention (CDC). Yellow Fever Vaccine Booster Doses: Recommendations of the Advisory Committee on Immunization Practices, 2015. MMWR Morb Mortal Wkly Rep. 2015 Jun 19. 64 (23):647-50. [View Abstract]
  71. Bilgic D, Daglar G, Sabanciogullari S, Ozkan SA. Attitudes of midwifery and nursing students in a Turkish university toward lesbians and gay men and opinions about healthcare approaches. Nurse Educ Pract. 2018 Mar. 29:179-184. [View Abstract]
  72. Daddario-DiCaprio KM, Geisbert TW, Geisbert JB, Stroher U, Hensley LE, Grolla A, et al. Cross-protection against Marburg virus strains by using a live, attenuated recombinant vaccine. J Virol. 2006 Oct. 80(19):9659-66. [View Abstract]
  73. Martin JE, Sullivan NJ, Enama ME, Gordon IJ, Roederer M, Koup RA, et al. A DNA vaccine for Ebola virus is safe and immunogenic in a phase I clinical trial. Clin Vaccine Immunol. 2006 Nov. 13(11):1267-77. [View Abstract]

New World Arenavirus - Machupo. Oral mucosal hyperemia and hemorrhage in a patient with Bolivian hemorrhagic fever.

Bunyavirus infection. Ecchymoses encompassing left upper extremity one week after onset of CCHF. Ecchymoses often are accompanied by hemorrhage in other locations: epistaxis, puncture sites, hematemesis, melena, and hematuria. Image provided by Robert Swaneopoel, PhD, DTVM, MRCVS, National Institute of Virology, Sandringham, South Africa.

Filovirus disease - Ebola fever. Patient with Ebola hemorrhagic fever during 1976 outbreak in Zaire demonstrating palatal petechiae and hemorrhage. Photo courtesy of Joel Breman.

Patient with morbilliform exanthem of dengue fever. Note islands of sparing characteristics for dengue. Photo courtesy Duane Gubler, PhD.

New World Arenavirus - Machupo. Oral mucosal hyperemia and hemorrhage in a patient with Bolivian hemorrhagic fever.

Bunyavirus infection. Ecchymoses encompassing left upper extremity one week after onset of CCHF. Ecchymoses often are accompanied by hemorrhage in other locations: epistaxis, puncture sites, hematemesis, melena, and hematuria. Image provided by Robert Swaneopoel, PhD, DTVM, MRCVS, National Institute of Virology, Sandringham, South Africa.

Filovirus disease - Ebola fever. Patient with Ebola hemorrhagic fever during 1976 outbreak in Zaire demonstrating palatal petechiae and hemorrhage. Photo courtesy of Joel Breman.

Patient with morbilliform exanthem of dengue fever. Note islands of sparing characteristics for dengue. Photo courtesy Duane Gubler, PhD.

Virus Family and Genus Type of Hemorrhagic Fever Reservoir Host Geographic Distribution
Arenaviridae



Guanarito



Junin



Machupo



Lassa



Sabia



 



Venezuelan



Argentinian



Bolivian



Lassa (West Africa)



Brazilian or Sao Paulo



 



Rodents



 



Venezuela



Argentina



Bolivia



West Africa



Brazil



Bunyaviridae



Nairovirus



Phlebovirus



Hantaan virus



 



Crimean-Congo



Rift Valley



Korean



HPS



 



Ticks



Mosquito and contact with infected blood in slaughter houses



Contact with infected rodents and their excreta



Crimea, Central Africa, South Africa, Iraq, Pakistan, Turkey, Iran, Afghanistan, Russia



Africa, Egypt



Korea, Eastern Europe, Russia, Scandinavia



North, Central, and South America



Flaviviridae



Flavivirus



Flavivirus



Flavivirus



Flavivirus



 



Yellow



Dengue



Chikungunya



Omsk



 



Mosquito



Mosquito



Mosquito



Tick



 



Tropical Africa, South America



Entire tropical zone



India, Southeast Asia



Siberia



Filoviridae



Marburg



Ebola



 



Marburg



Ebola



 



Infected monkeys were implicated but no known definite reservoir



 



Africa



West Africa



Virus Family and Type of VHF Mortality Rate, %
Arenaviridae



Argentinian and Bolivian



Lassa (West African)



Venezuelan and Sao Paulo



 



10-30



30-40



33



Bunyaviridae



Korean and Seoul



Rift Valley



Congo-Crimean



HPS



 



5-15



1



10-50



15-50



Flaviviridae



Yellow



Dengue



 



< 1



5



Filoviridae



Marburg



Ebola



 



23-25



25-100