MOBILE VIEW  | 

FILOVIRUS INFECTIONS

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Filoviruses, in the family Filoviridae, are causative agents of a hemorrhagic fever in man, with mortality rates ranging from 22% to 88%. These viruses are enveloped, nonsegmented negative-stranded RNA viruses, which are zoonoses, and are separated into two types: Marburg and Ebola. Person-to person transmission by intimate contact with body fluids is the main route of infection in human filoviral hemorrhagic fever epidemics, with an incubation period ranging from 2 to 21 days, and averaging one week. Currently, the natural reservoir and natural mode of transmission for filoviruses are unknown. All forms of the viral hemorrhagic fever begin with fever and muscle aches. Monkeys, as well as humans, appear to be susceptible to infection and may serve as a source of virus if infected.

Specific Substances

    A) EBOLA VIRUS DISEASE
    1) Ebola
    2) Ebola hemorrhagic fever
    3) Ebola/Bundibugyo
    4) Ebola/Cote d'Ivoire
    5) Ebola/Reston
    6) Ebola/Sudan
    7) Ebola/Tai forest
    8) Ebola/Zaire
    MARBURG VIRUS DISEASE
    1) Marburg
    2) Marburg hemorrhagic fever
    3) Marburg/Musoki
    4) Marburg/Popp
    5) Marburg/Ravn
    GENERAL TERMS
    1) African hemorrhagic fever
    2) Marburg-Ebola disease
    3) Filoviridae

Available Forms Sources

    A) FORMS
    1) Filoviruses, belonging to the family, Filoviridae, are classified as biosafety level 4 agents, limiting work with these agents to a few selected laboratories (Feldman et al, 1996). These viruses are enveloped, nonsegmented negative-stranded RNA viruses, separated into two types: Marburg and Ebola. Marburg and Ebola are serologically, biochemically and genetically distinct from one another. Little genetic variability occurs among viruses of the Marburg type
    a) EBOLA
    1) Ebola Virus Disease is a rare, potentially fatal, disease caused by an infection of an Ebola virus strain. Ebola Virus Disease has also been called Ebola hemorrhagic fever. Four of the 5 identified Ebola virus species cause disease in humans: Ebola virus (Zaire ebolavirus), Sudan virus (Sudan ebolavirus), Tai Forest virus (Tai Forest ebolavirus, formerly Cote d'lvoire ebolavirus), and Bundibugyo virus (Bundibugyo ebolavirus). Reston virus (Reston ebolavirus) is the fifth virus species that has caused disease in nonhuman primates only. Outbreaks of Ebola have developed sporadically in Africa since it was discovered in 1976 near the Ebola River (Centers for Disease Control and Prevention (CDC), 2014; Breman et al, 1997; Le Guenno et al, 1995).
    2) The Zaire subtype of Ebola appears most virulent to humans, while the Reston subtype has caused infection, but not disease in 4 humans (CDC, 1990). McCormick et al (1983) reported that fewer than 10 infectious particles of a Zaire strain were lethal for mice, whereas 10,000 infectious particles of a Sudan strain were unable to kill any of the mice (McCormick et al, 1983).
    3) Ebola virus is pleomorphic and filamentous, with an average length of 920 nm and average diameter of 80 nm. The virus is a linear, single stranded RNA molecule, enclosed in a helical capsid, and covered by an envelope containing virus-specified glycoprotein spikes (Al-Ahdal, 1995). Ebola virus is structurally similar to Marburg virus, but antigenically different.
    b) MARBURG
    1) Johnson et al (1996) have described 3 strains or subtypes of Marburg virus: Ravn (RAV), Musoki (MUS) and Popp (POP) (Johnson et al, 1996).
    2) A potential for evolution of the filoviruses exists, due to an error rate of the virus-encoded polymerase and lack of repair mechanisms, exists. Consequences of evolution include genetic variants which are selected by the hosts for different transmissibility, virulence and other biological properties. However, sequence analysis of filovirus isolates has suggested a low frequency for emergence of variants. Mutant virus populations and the probability of a filovirus emerging as a serious public health problem could occur due to increases in human population, increases in speed, variety and frequency of travel, and disruption of social structures (Feldman et al, 1996).
    B) SOURCES
    1) The natural reservoir of filoviruses, where it propagates between human attacks, is unknown. Non-human primates are infected by filoviruses and may transmit the disease to humans (Al-Ahdal, 1995).
    2) EBOLA: The natural reservoir of the Ebola virus is unknown. The most likely reservoir is bats; 4 of the 5 virus strains are found in an animal host native to Africa (Centers for Disease Control and Prevention (CDC), 2014).
    C) USES
    1) Use of Ebola virus as a biological warfare agent has been proposed due to its highly contagious and lethal properties. However, because of uncertain stability outside of animal hosts, it may not be desirable as a biological agent for warfare unless a stable aerosolized form is developed (Cole, 1996).

Life Support

    A) This overview assumes that basic life support measures have been instituted.

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) TOXIC CLASS: The family Filoviridae, which consists of a single genus, filovirus, is separated into two types: Marburg and Ebola. They are nonsegmented negative-stranded RNA viruses. Filoviruses are classified as biosafety level 4 agents, limiting work with these agents to a few selected laboratories.
    B) TOXICOLOGY: Filoviruses replicate in endothelial cytoplasm causing focal necrosis, separation of tight junctions, and basement membrane detachment. Following viral replication in many organs, focal necrosis has been reported to occur in the liver, spleen, kidneys, lungs, testis, and lymphatics, with widespread vascular damage due to impaired microcirculation. The blood fails to clot, and main lesions appear in the vascular endothelium and platelets, resulting in severe bleeding. Disseminated intravascular coagulation (DIC) and thrombocytopenia are common.
    C) TRANSMISSION: Transmission occurs via direct contact with body fluids; during the critical or terminal stages of illness, high viral titers render body fluids highly infectious. Nosocomial transmission has been reported. Monkeys, humans, other primates, and bats appear to be susceptible to infection and may serve as a source of virus if infected.
    D) EPIDEMIOLOGY: EBOLA: In the 2014 Ebola Outbreak in West Africa, countries with widespread transmission were Guinea, Liberia, and Sierra Leone. Nigeria had localized transmission, and Senegal and the United States had travel-associated cases. MARBURG: Marburg virus is indigenous to Africa. Outbreaks have rarely been reported.
    E) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: After the incubation period, abrupt initial symptoms may occur, including influenza-like syndrome, fever, headache, myalgia, joint pain, and sore throat. This is commonly followed by vomiting, diarrhea, and abdominal pain. A transient, morbilliform and desquamating skin rash often appears 5 to 7 days later.
    2) SEVERE TOXICITY: Beginning the second week of the illness, severe hemorrhagic manifestations are common. Gastrointestinal bleeding, which can arise from any part of the GI tract, is most common, although bleeding may occur from other organ sites, any orifice, mucous membranes, and skin. Disseminated intravascular coagulation and thrombocytopenia are common. Impaired renal and hepatic function may occur. A short normothermic period sometimes precedes multiorgan failure and fatal shock.
    0.2.3) VITAL SIGNS
    A) WITH POISONING/EXPOSURE
    1) Abrupt onset of fever, which is persistent throughout the illness, is common.
    2) Decreased blood pressure is common following fluid losses.
    0.2.20) REPRODUCTIVE
    A) Of 73 pregnant fatalities from an Ebola virus outbreak in Zaire, 25% experienced abortions prior to death.

Laboratory Monitoring

    A) Obtain CBC, electrolytes, PT and PTT, renal function tests, liver enzymes, blood cultures, CSF, ascites fluid, or pleural fluid as clinically indicated. Because filoviruses are classified as biosafety level 4 agents, work with these agents is limited to a few selected laboratories, such as the CDC in Atlanta, Georgia.
    B) Enzyme-linked immunosorbent assay (ELISA) and electrophoretic immunotransblot test (EITB, Western blot) may be valuable serologic diagnostic tests when performed in selected laboratories.
    C) Ebola virus is usually detectable in blood by real-time polymerase chain reaction testing 3 to 10 days post-symptom onset.

Treatment Overview

    0.4.3) INHALATION EXPOSURE
    A) Filoviruses are not known to be transmitted from human-to-human via inhalation.
    0.4.4) EYE EXPOSURE
    A) Mucocutaneous exposures to blood, body fluids, secretions, or excretions from patients with suspected filovirus infections should be immediately rinsed with copious amounts water or eyewash solution.
    0.4.5) DERMAL EXPOSURE
    A) OVERVIEW
    1) MANAGEMENT OF MILD TO MODERATE TOXICITY
    a) Intensive supportive treatment is the mainstay of therapy. Correct any significant fluid and/or electrolyte abnormalities in patients with severe diarrhea and/or vomiting.
    2) MANAGEMENT OF SEVERE TOXICITY
    a) Intensive supportive treatment is the mainstay of therapy. Transfusions of fresh frozen plasma/packed red blood cells, and platelets (as needed) for severe thrombocytopenia, bleeding. Treat severe hypotension with IV 0.9% NaCl at 10 to 20 mL/kg. Add dopamine or norepinephrine if unresponsive to fluids. Hemodialysis may be necessary in cases of renal failure.
    3) DECONTAMINATION
    a) PREHOSPITAL: Remove contaminated clothing immediately. Sites exposed to blood/body fluids should be immediately washed with soap and water. FIRST RESPONDERS: Any healthcare personnel should follow strict isolation precautions (ie, includes gloves, gown, mask) and avoid direct contact with blood or bodily fluids of a patient with suspected Ebola or Marburg virus infection.
    b) HOSPITAL: Strict barrier techniques and infection control are recommended while treating any patient with confirmed or suspected Ebola or Marburg virus infection.
    4) AIRWAY MANAGEMENT
    a) In cases of airway compromise, supportive measures including endotracheal intubation and mechanical ventilation may be necessary.
    5) ANTIDOTE
    a) No antidote or vaccine is available for filovirus infections.
    6) ENHANCED ELIMINATION PROCEDURE
    a) Hemodialysis is unlikely to be necessary, but may be needed in patients that develop acute renal failure.
    7) PATIENT DISPOSITION
    a) HOME CRITERIA: Close contacts of filovirus-infected patients should be stringently monitored and asked to report any fever (ie, record temperature twice daily for 3 weeks). Surveillance should be continued for 3 weeks after the date of the last contact.
    b) OBSERVATION CRITERIA: Any person who has a history of close physical contact with an infected patient(s) should be put under strict surveillance (ie, obtain temperature twice daily; in the case of a temperature greater than 38.3 degrees C (101 degrees F), hospitalize the patient immediately and place in strict isolation).
    c) ADMISSION CRITERIA: All patients with suspected filovirus infections should be admitted to a strict isolation unit that can provide intensive care as needed; the staff should use barrier techniques.
    d) CONSULT CRITERIA: Infectious disease staff/intensivists, should manage the care of these patients. All confirmed and suspected cases should be reported immediately through local and State health departments to the Viral Special Pathogens Branch of the Centers for Disease Control (CDC).
    8) PITFALLS
    a) Failure to diagnose filovirus infection. Failure to appropriately use barriers or personal protective equipments causing further infection and exposure. Failure to recognize shock and hemorrhage.
    9) DIFFERENTIAL DIAGNOSIS
    a) Includes other hemorrhagic fevers (eg, Crimean-Congo hemorrhagic fever, Dengue hemorrhagic fever), typhoid fever, meningococcemia, toxic shock syndrome, malaria. Also includes noninfectious causes of bleeding manifestations, (eg, idiopathic or thrombotic thrombocytopenic purpura, hemolytic uremic syndrome, acute leukemia, and collagen-vascular diseases).

Range Of Toxicity

    A) TOXICITY: Animal experiments have shown fatal inhalation doses of Ebola virus to be 400 plaque forming units of virus.

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) ALTERED MENTAL STATUS
    1) WITH POISONING/EXPOSURE
    a) After about a week of illness, mental status changes often occur and include confusion, lack of concentration, memory loss, and delirium (Edmond et al, 1977; Johnson et al, 1996; Sodhi, 1996).
    b) Delirium may be persistent throughout the infectious course of illness (Al-Ahdal, 1995; WHO, 1978a). Combativeness and bizarre behavior were commonly seen during a 1976 Ebola outbreak in Sudan (n=183), with abnormal behavior often persisting for several weeks following recovery (WHO, 1978a).
    B) HEADACHE
    1) WITH POISONING/EXPOSURE
    a) An early and common symptom of illness is severe headache and general malaise with chills and fever (Centers for Disease Control and Prevention (CDC), 2014; Geisbert & Jaax, 1998; CDC, 1988; WHO, 1995).
    1) INCIDENCE: In a 1976 Ebola outbreak in Sudan, 100% of patients (n=183) experience headaches (WHO, 1978a).
    C) CENTRAL NERVOUS SYSTEM FINDING
    1) WITH POISONING/EXPOSURE
    a) It is not unusual for sudden blindness, tinnitus, hearing loss, and dysesthesias to develop in the late phases of filovirus infections (Bwaka et al, 1999; Rollin & Ksiazek, 1998).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) GASTROINTESTINAL HEMORRHAGE
    1) WITH POISONING/EXPOSURE
    a) Beginning the second week of the illness, frank bleeding, which can arise from any part of the gastrointestinal tract and from multiple other sites, is common (Centers for Disease Control and Prevention (CDC), 2014; CDC, 1988; Geisbert & Jaax, 1998; WHO, 1978b; WHO, 1995; Johnson et al, 1996).
    b) Gingival bleeding may occur (WHO, 1978b; Baron et al, 1983; Anon, 1996; Johnson et al, 1996).
    B) NAUSEA, VOMITING AND DIARRHEA
    1) WITH POISONING/EXPOSURE
    a) Early in the illness, anorexia, nausea, abdominal pain, vomiting and diarrhea often occur, and may be persistent, resulting in significant fluid losses (Centers for Disease Control and Prevention (CDC), 2014; Leroy et al, 2002; CDC, 1988; Edmond et al, 1977; Geisbert & Jaax, 1998; WHO, 1978b; Al-Ahdal, 1995; Baron et al, 1983; Rollin & Ksiazek, 1998; Bwaka et al, 1999; Formenty et al, 1999).
    1) Diarrhea and vomitus is often bloody (Leroy et al, 2002).
    2) INCIDENCE: Diarrhea and vomiting have been reported in 81% and 59% of patients (n=183), respectively, in a 1976 Ebola outbreak in Sudan (WHO, 1978a). Melena was reported in 59% of these patients.
    C) PANCREATITIS
    1) WITH POISONING/EXPOSURE
    a) Acute pancreatitis may be part of the pathogenesis of Ebola hemorrhagic fever (WHO, 1978b).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) TOXIC HEPATITIS
    1) WITH POISONING/EXPOSURE
    a) Non-icteric hepatitis is included in the pathogenesis of filoviral infections (WHO, 1978b). Serum liver function tests may be markedly elevated. Focal necrosis of the liver has been reported (Formenty et al, 1999; Al-Ahdal, 1995).
    3.9.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) HEPATIC ENZYMES INCREASED
    a) MONKEYS: Abrupt, marked elevation of hepatic enzymes, including LDH, ALT and AST, were reported in cynomolgus monkeys with Ebola viral infections (Dalgard et al, 1992).
    2) HEPATIC NECROSIS
    a) MONKEYS: Histopathologic examination of monkeys infected with Ebola Reston virus revealed hepatocellular necrosis ranging from individual cells to clusters multifocally scattered throughout the parenchyma. This appeared to be a common effect in the infected monkeys (Dalgard et al, 1992).
    3) SPLENOMEGALY
    a) MONKEYS: Splenomegaly was the most consistent finding on necropsy in monkeys with known Ebola Reston virus infection. The spleen was often 3 to 4 times normal size (Dalgard et al, 1992).

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) ALBUMINURIA
    1) WITH POISONING/EXPOSURE
    a) Proteinuria has been reported; it was noted on the fourth day of illness and persisted for about one week (Edmond et al, 1977). Decreased urine output was also noted.
    b) Proteinuria was reported as uniformly positive and was used as a major diagnostic criterion in an Ebola hemorrhagic fever outbreak in Zaire in 1976 (WHO, 1978b). Proteinuria was observed in a few patients in a 1976 Ebola outbreak in Sudan (WHO, 1978a).
    B) RENAL FAILURE SYNDROME
    1) WITH POISONING/EXPOSURE
    a) Oliguria and anuria have been reported following filoviral infections (WHO, 1978b). In severe cases renal failure, necessitating hemodialysis, may occur (Johnson et al, 1996).
    C) BLOOD IN URINE
    1) WITH POISONING/EXPOSURE
    a) Multifocal petechial hemorrhages have been observed in the renal cortex and pelvis, and urinary bladder at necropsy (Geisbert & Jaax, 1998).
    1) INCIDENCE: In a 1976 Ebola outbreak in Sudan (n=183), gross hematuria was seldom seen (WHO, 1978a).
    D) SEMEN EXAM: ABNORMAL
    1) WITH POISONING/EXPOSURE
    a) Eighteen days following exposure to Ebola virus from an accidental needle prick, semen was still positive for the virus. The patient recovered (Edmond et al, 1977).
    b) A seminal fluid sample, obtained from one patient who was recovering from an Ebola virus infection during the 1995 Ebola hemorrhagic fever outbreak in Kikwit, Democratic Republlic of Congo, was positive for the infectious virus 82 days post-onset of the disease (Rodriguez et al, 1999).
    E) ORCHITIS
    1) WITH POISONING/EXPOSURE
    a) Orchitis was occasionally manifested in males during a 1976 Ebola outbreak in Sudan (WHO, 1978a). Unilateral orchitis is reported as a late manifestation of disease in convalescent patients (Bwaka et al, 1999).

Summary Of Exposure

    A) TOXIC CLASS: The family Filoviridae, which consists of a single genus, filovirus, is separated into two types: Marburg and Ebola. They are nonsegmented negative-stranded RNA viruses. Filoviruses are classified as biosafety level 4 agents, limiting work with these agents to a few selected laboratories.
    B) TOXICOLOGY: Filoviruses replicate in endothelial cytoplasm causing focal necrosis, separation of tight junctions, and basement membrane detachment. Following viral replication in many organs, focal necrosis has been reported to occur in the liver, spleen, kidneys, lungs, testis, and lymphatics, with widespread vascular damage due to impaired microcirculation. The blood fails to clot, and main lesions appear in the vascular endothelium and platelets, resulting in severe bleeding. Disseminated intravascular coagulation (DIC) and thrombocytopenia are common.
    C) TRANSMISSION: Transmission occurs via direct contact with body fluids; during the critical or terminal stages of illness, high viral titers render body fluids highly infectious. Nosocomial transmission has been reported. Monkeys, humans, other primates, and bats appear to be susceptible to infection and may serve as a source of virus if infected.
    D) EPIDEMIOLOGY: EBOLA: In the 2014 Ebola Outbreak in West Africa, countries with widespread transmission were Guinea, Liberia, and Sierra Leone. Nigeria had localized transmission, and Senegal and the United States had travel-associated cases. MARBURG: Marburg virus is indigenous to Africa. Outbreaks have rarely been reported.
    E) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: After the incubation period, abrupt initial symptoms may occur, including influenza-like syndrome, fever, headache, myalgia, joint pain, and sore throat. This is commonly followed by vomiting, diarrhea, and abdominal pain. A transient, morbilliform and desquamating skin rash often appears 5 to 7 days later.
    2) SEVERE TOXICITY: Beginning the second week of the illness, severe hemorrhagic manifestations are common. Gastrointestinal bleeding, which can arise from any part of the GI tract, is most common, although bleeding may occur from other organ sites, any orifice, mucous membranes, and skin. Disseminated intravascular coagulation and thrombocytopenia are common. Impaired renal and hepatic function may occur. A short normothermic period sometimes precedes multiorgan failure and fatal shock.

Vital Signs

    3.3.1) SUMMARY
    A) WITH POISONING/EXPOSURE
    1) Abrupt onset of fever, which is persistent throughout the illness, is common.
    2) Decreased blood pressure is common following fluid losses.
    3.3.3) TEMPERATURE
    A) WITH POISONING/EXPOSURE
    1) Sudden onset of chills and high fever (greater than 38.6 degrees C), after a usual incubation period of 5 to 10 days, is characteristic of filovirus infections. Fever is persistent throughout the illness (Centers for Disease Control and Prevention (CDC), 2014; WHO, 1978a; WHO, 1978b; WHO, 1995; Geisbert & Jaax, 1998; Edmond et al, 1977; CDC, 1988; Shope, 1996; Feldman et al, 1996; Anon, 1996; Rollin & Ksiazek, 1998).
    a) INCIDENCE: During a 1976 Ebola outbreak in Sudan, 100% of patients (n=183) experienced fever (WHO, 1978a).
    3.3.4) BLOOD PRESSURE
    A) WITH POISONING/EXPOSURE
    1) Hypotension may commonly be noted in hemorrhagic viral infections (Shope, 1996; Halstead, 1996).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) HEMORRHAGE
    1) WITH POISONING/EXPOSURE
    a) Beginning the second week of the illness, severe hemorrhagic manifestations are common. Gastrointestinal bleeding, which can arise from any part of the GI tract, is most common, although bleeding may occur from other organ sites, any orifice, mucous membranes, and skin (Centers for Disease Control and Prevention (CDC), 2014; Geisbert & Jaax, 1998; CDC, 1988; WHO, 1978b; Sodhi, 1996). Overt hemorrhagic features may supervene from day 5 onward (Richards et al, 2000). Hemoglobin levels will be decreased during the acute phase of illness during active hemorrhaging. Richards et al (2000) reported a death due to refractory thrombocytopenia and an intracerebral hemorrhage in one patient (Richards et al, 2000).
    1) INCIDENCE: Hemorrhaging was reported in 71% of patients (n=183) in a 1976 Ebola outbreak in Sudan. Of the fatal cases, 91% exhibited bleeding, and of the recovered cases, 48% exhibited bleeding (WHO, 1978a).
    b) Following viral replication in many organs, focal necrosis occurs in the liver, kidneys, and lymphatics. The blood fails to clot, and main lesions appear in the vascular endothelium and the platelets, resulting in severe bleeding in the liver, mucosa, abdomen, pericardium, and other organs.
    B) DISSEMINATED INTRAVASCULAR COAGULATION
    1) WITH POISONING/EXPOSURE
    a) Disseminated intravascular coagulation (DIC) may occur in the later stages of illness and is usually present in fatal cases. Thrombocytopenia, prolonged prothrombin, and partial thromboplastin times are reported to be consistent with DIC (Geisbert & Jaax, 1998; WHO, 1978b; Johnson et al, 1996; CDC, 1995a; Borio et al, 2002). DIC may lead to micro-thrombi with surrounding tissue infarction (Johnson et al, 1996). Thrombocytopenia may be evident early in the disease; a petechial rash may appear on days 3-10 (Richards et al, 2000).
    C) LEUKOCYTOSIS
    1) WITH POISONING/EXPOSURE
    a) Leukocytosis may occur in filovirus infections (Geisbert & Jaax, 1998; Johnson et al, 1996). White blood cell counts, when measured, were elevated in patients following Ebola viral infections in Sudan in 1976 (WHO, 1978a).
    D) LEUKOPENIA
    1) WITH POISONING/EXPOSURE
    a) Neutrophilia and lymphopenia are striking features of filoviral infections and occur early in the illness (CDC, 1988; Edmond et al, 1977; Takada & Kawaoka, 1998; Sodhi, 1996).
    1) White blood cell counts were depressed and did not return to normal until 3 months after the onset of an Ebola virus infection (Edmond et al, 1977).
    3.13.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) LEUKOPENIA
    a) GUINEA PIGS: Four days following aerogenic infection with Marburg virus, guinea pigs exhibited leukopenia and hyperthermia as the earliest virological and clinical signs of disease (Lub et al, 1995).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) MACULOPAPULAR ERUPTION
    1) WITH POISONING/EXPOSURE
    a) Approximately 5 to 7 days after the onset of illness, a transient and morbilliform skin rash, which subsequently desquamates, often appears (WHO, 1978b; Edmond et al, 1977; CDC, 1988; Johnson et al, 1996; Rollin & Ksiazek, 1998; Bwaka et al, 1999; Formenty et al, 1999; Borio et al, 2002). The rash often spreads to all areas of the body and may become confluent.
    1) INCIDENCE: Rash or desquamation was reported in 52% of patients (n=183) in a 1976 Ebola outbreak in Sudan (WHO, 1978a).
    B) PURPURA
    1) WITH POISONING/EXPOSURE
    a) Beginning the second week of the illness, hemorrhagic manifestations appear and include petechiae as well as frank bleeding (Centers for Disease Control and Prevention (CDC), 2014; CDC, 1988; Johnson et al, 1996; Sodhi, 1996; Edmond et al, 1977).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) JOINT PAIN
    1) WITH POISONING/EXPOSURE
    a) Severe malaise, extreme weakness with myalgias, and joint pain are commonly noted throughout the course of illness (Centers for Disease Control and Prevention (CDC), 2014; CDC, 1988; Edmond et al, 1977; WHO, 1978b; WHO, 1995; Al-Ahdal, 1995; Rollin & Ksiazek, 1998). Asthenia may persist for several weeks following recovery (Rollin & Ksiazek, 1998). Arthralgia, asymmetrical or symmetrical, occurs. It is sometimes migratory, principally involves the large joints, and is a common late manifestation of disease in convalescent patients (Bwaka et al, 1999; Rowe et al, 1999).
    b) CASE REPORT: A woman from Medemba village in Gabon presented to the hospital with symptoms of high fever, asthenia, arthralgia, diarrhea, and vomiting. The patient died 4 days later, with evidence of blood in her feces and mouth. She was the fifth person in her family to develop similar symptoms and subsequently die. The symptoms and inter-family transmission suggested Ebola hemorrhagic fever. Sera from two patients, who had developed similar symptoms and had died after coming into contact with the woman, showed high titers of Ebola virus antigen and contained Ebola virus RNA, confirming the suspected diagnosis of Ebola hemorrhagic fever (Leroy et al, 2002).

Endocrine

    3.16.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) ADRENAL INSUFFICIENCY
    a) MONKEYS: Multifocal necrosis of the zona glomerulosa in the adrenal cortex was noted on histopathology examination in monkeys infected with Ebola Reston virus (Dalgard et al, 1992).

Immunologic

    3.19.2) CLINICAL EFFECTS
    A) INFECTIOUS DISEASE
    1) WITH POISONING/EXPOSURE
    a) Asymptomatic, replicative Ebola infection can occur in humans. Seroconversion occurs, however, circulating Ebola antigen is not detected. It is suggested that asymptomatic infections do not result from viral mutations. These individuals exhibit a strong inflammatory response early in the infectious process, as characterized by high circulating concentrations of cytokines and chemokines (Leroy et al, 2000).
    b) Due to immunosuppression, superinfections with bacteria or fungus may occur during the course of a filoviral infection. Johnson et al (1996) and Geisbert & Jaax (1998) reported Pseudomonas aeruginosa isolated from the blood of fatal Marburg hemorrhagic fever victims (Johnson et al, 1996; Geisbert & Jaax, 1998), and Edmond et al (1977) reported an extensive candidiasis in throat tissue of an Ebola hemorrhagic fever victim (Edmond et al, 1977).
    c) Infections with Klebsiella, E coli and S aureus have also been reported (Geisbert & Jaax, 1998). Secondary bacteremia and sepsis may develop due to damage of the gastrointestinal mucosa.
    B) LYMPHADENOPATHY
    1) WITH POISONING/EXPOSURE
    a) Lymphadenopathy may be seen during the acute phase of filoviral illnesses (Geisbert & Jaax, 1998; Sodhi, 1996).
    C) DISORDER OF IMMUNE FUNCTION
    1) WITH POISONING/EXPOSURE
    a) A significant level of immune activation accompanies and potentially contributes to terminal Ebola virus infections. Markedly elevated levels of interferon (IFN)-y, IFN-a, interleukin (IL)-2, IL-10, and tumor necrosis factor-a are associated with fatal Ebola viral infections (Villinger et al, 1999).

Reproductive

    3.20.1) SUMMARY
    A) Of 73 pregnant fatalities from an Ebola virus outbreak in Zaire, 25% experienced abortions prior to death.
    3.20.3) EFFECTS IN PREGNANCY
    A) PLACENTAL BARRIER
    1) The World Health Organization determined that infants born to patients with suspected filovirus hemorrhagic fever were called neonatal cases if they died within 28 days of birth. All of 11 live infants born to mothers who died of hemorrhagic fever died within 19 days. Seven of the infants had fevers, but bleeding was infrequent (WHO, 1978b). Virological and pathological data were absent in these 11 cases.
    B) ABORTION
    1) Of 73 pregnant fatalities from an Ebola virus outbreak in Zaire, 25% experienced abortions prior to death (WHO, 1978b). Premature labor occasionally occurred in pregnant females during a 1976 Ebola outbreak in Sudan (WHO, 1978a). Mupapa et al (1999a) reported 66% of 15 pregnant women with Ebola hemorrhagic fever had spontaneous abortions. Mortality among pregnant women was not significantly higher than the overall mortality observed during an Ebola epidemic.

Heent

    3.4.3) EYES
    A) WITH POISONING/EXPOSURE
    1) Conjunctivitis may occur during the first week of illness and is characteristic of viral hemorrhagic fevers (CDC, 1988; Anon, 1996).
    2) Late manifestations of disease occur in convalescent patients and can include ocular diseases (conjunctivitis, unilateral loss of vision, uveitis) (Bwaka et al, 1999; Kibadi et al, 1999).
    3) A periorbital mucormycosis abscess on eyelid tissue developed in one patient. Following convalescence, the patient had severe sequelae associated with the mucormycosis (Kalongi et al, 1999).
    3.4.4) EARS
    A) WITH POISONING/EXPOSURE
    1) Tinnitus and hearing loss were recorded more often in the Kikwit Ebola outbreak in 106 patients (Breman et al, 1997) than in previous outbreaks, and are not unusual in the late phase of the disease (Bwaka et al, 1999; Rollin & Ksiazek, 1998).
    3.4.5) NOSE
    A) WITH POISONING/EXPOSURE
    1) Epistaxis was frequently observed during a 1976 Ebola outbreak in Sudan (n=183) (WHO, 1978a).
    3.4.6) THROAT
    A) WITH POISONING/EXPOSURE
    1) Progressive sore throat and pharyngitis, which is frequently exudative, is common in the early stages of filovirus illness (4 to 7 days after onset of illness) (Bwaka et al, 1999; CDC, 1988; Edmond et al, 1977; WHO, 1978b; WHO, 1995; Sodhi, 1996).
    a) Edema of the soft palate and pharynx may lead to dysphagia. Cough may be associated with oral/throat lesions. Aphthous-like ulcers may be seen in the oral cavities in some patients (WHO, 1978a).
    b) INCIDENCE: Sore throat and cough were reported in 63% and 49% of patients (n=183), respectively, in a 1976 Ebola outbreak in Sudan (WHO, 1978a).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) HYPOTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) Hypotension is commonly seen during the acute stage of illness. Death may ensue as a result of intractable hemorrhagic shock and multiple organ failure (Geisbert & Jaax, 1998; Johnson et al, 1996; Sodhi, 1996) (CDC, 1995).
    b) In 3 cases of Ebola infection, hypovolemic shock preceded death (Baron et al, 1983). Death may be due to cardiac failure (Johnson et al, 1996). Clinical shock, in severe cases, may lead to diffuse coagulative necrosis (Johnson et al, 1996).
    B) CHEST PAIN
    1) WITH POISONING/EXPOSURE
    a) Chest pain is a common symptom of Ebola viral infections, reported in 83% of patients (n=183) in a 1976 Ebola outbreak in Sudan (WHO, 1978a).
    b) Chest pain was often associated with dry cough. Basilar rales were commonly noted on chest examination. The incidence of chest pain in Marburg viral infections appears to be less frequent (WHO, 1978a).
    C) TACHYARRHYTHMIA
    1) WITH POISONING/EXPOSURE
    a) Two patients with Ebola hemorrhagic fever died with terminal tachycardia (WHO, 1978b).
    D) ELECTROCARDIOGRAM ABNORMAL
    1) WITH POISONING/EXPOSURE
    a) ECG tracings suggestive of myocarditis and pericarditis have been described in patients with filovirus diseases (WHO, 1978a).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) HEMORRHAGE
    1) WITH POISONING/EXPOSURE
    a) Blood-tinged pleural effusions and hemorrhagic lungs and tracheobronchial tree have been reported on autopsy following filoviral illnesses (Geisbert & Jaax, 1998; Johnson et al, 1996). Focal necrosis is widely seen in the lungs (Sodhi, 1996).
    B) HYPERVENTILATION
    1) WITH POISONING/EXPOSURE
    a) Hiccup and tachypnea may occur in severe cases and are usually precursors of death (Rollin & Ksiazek, 1998). Hiccups were reported in 15% of Ebola viral cases (n=66) in a Kikwit (area of Zaire) outbreak (CDC, 1995b). A high frequency (25%) of hyperventilation was reported as a terminal event, probably reflecting underlying metabolic abnormalities rather than cardiac or lung involvement (Bwaka et al, 1999).
    3.6.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) RESPIRATORY DISORDER
    a) GUINEA PIGS: Following aerogenic infection with Marburg virus, primary viral multiplication occurred in the lungs and was reported at the initial stage of infection. Bronchopulmonary washings revealed the presence of virus 2 to 3 days after the infection (Lub et al, 1995).
    2) INTERSTITIAL PNEUMONIA
    a) MONKEYS: Patchy interstitial pneumonia was noted on histopathology examination of monkeys infected with Ebola Reston virus (Dalgard et al, 1992).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Obtain CBC, electrolytes, PT and PTT, renal function tests, liver enzymes, blood cultures, CSF, ascites fluid, or pleural fluid as clinically indicated. Because filoviruses are classified as biosafety level 4 agents, work with these agents is limited to a few selected laboratories, such as the CDC in Atlanta, Georgia.
    B) Enzyme-linked immunosorbent assay (ELISA) and electrophoretic immunotransblot test (EITB, Western blot) may be valuable serologic diagnostic tests when performed in selected laboratories.
    C) Ebola virus is usually detectable in blood by real-time polymerase chain reaction testing 3 to 10 days post-symptom onset.
    4.1.2) SERUM/BLOOD
    A) BLOOD/SERUM CHEMISTRY
    1) Obtain CBC, electrolytes, renal function tests, liver enzymes, blood cultures, CSF, ascites fluid, or pleural fluid as clinically indicated. Because filoviruses are classified as biosafety level 4 agents, work with these agents is limited to a few selected laboratories, such as the CDC in Atlanta, Georgia (Feldman et al, 1996). Only specially trained personnel should handle blood and other biologic specimens. Blood and autopsy specimens should be placed in tightly sealed metal containers, wrapped in absorbent material inside a sealed plastic bag, and shipped on dry ice to laboratories with BSL - 4 biocontainment facilities.
    2) Obtain specimens for testing when a symptomatic patient suspected of Ebola exposure reports to a healthcare facility. Ebola virus is detected in blood only after symptom onset, and it may take up to 3 days after onset of symptoms for the virus to reach detectable levels. The Ebola virus is usually detectable by real-time polymerase chain reaction testing 3 to 10 days after the symptoms appear. If a patient presents less than 3 days after symptom onset and the first test is negative, a later specimen may be necessary (Centers for Disease Control and Prevention (CDC), 2014).
    3) Definitive diagnosis of Ebola or Marburg viral infections may be accomplished by isolation of the virus from blood or demonstrating IgM or rising IgG antibodies by IFA (CDC, 1988).
    4.1.3) URINE
    A) URINALYSIS
    1) Urine output should be measured in severe cases (Edmond et al, 1977). Midstream ("clean catch") specimens of urine may be essential for virus isolation, and should be put in a plastic screw-cap container for submission to a biosafety level 4 facility (CDC, 1988).

Methods

    A) BIOASSAY
    1) Amplification of filoviral RNA from serum can be accomplished with reverse transcription-polymerase-chain reaction (RT-PCR) assays. This method is highly sensitive and allows the direct sequencing of the genomic fragment and definitive diagnosis of the presence of Marburg or the subtype of Ebola virus (Peters, 1996; CDC, 1995a).
    B) IMMUNOASSAY
    1) The antigen-capture enzyme-linked immunosorbent assay (ELISA) has been shown to be effective in diagnosing filoviruses and is the CDC standard for confirming clinical diagnosis (Breman et al, 1997; CDC, 1995a). Immunoglobulin (Ig)M-capture ELISA detects antibodies in recently convalescent patients and IgG-ELISA appears to have overcome the problems of nonspecificity in the indirect fluorescent antibody test (Ksiazek et al, 1999a; Peters, 1996). Immunofluorescence and ELISA become positive during the second week of illness (Shope, 1996).
    2) Immunofluorescent antibody assay (IFA) appears to be one of the most accurate immunoassays for detection of Ebola and Marburg virus (Kalter et al, 1995) (van der Groen & Elliot, 1982)(Tomera, 1997; Johnson et al, 1996). Definitive diagnosis of Ebola or Marburg viral disease may be accomplished by isolation of the virus from blood or demonstrating IgM or rising IgG antibodies by IFA (CDC, 1988).
    a) Lupton (1981) has shown that acetone-fixed infected cell antigens that were exposed for 2 hours to 60Co irradiation to inactivate Ebola virus were better than ultraviolet-treated preparations in the indirect IFA test. Antigenicity was equal and false-positive results did not exist (Lupton, 1981).
    3) Kalter et al (1995) have described a dot-immunobinding assay (DIA) for the detection of Ebola-Reston virus antibody. Sera from 30 human and non-human primates were tested by ELISA, IFA, and Western blotting. The latter assay was retested by DIA, with all positive Western blotting assays also positive using the DIA method (Kalter et al, 1995).
    4) Merzlikin et al (1995) described an enzyme immunoassay (EIA) test system for detection of Ebola virus antigens and antibodies in human serum and tissue homogenates. It has shown high resolution and reproducibility of results, and allows detection of antibodies to Ebola virus starting from days 8 to 9 of infection (Merzlikin et al, 1995).
    5) Reverse transcription-polymerase chain reaction (RT-PCR) has been demonstrated to be effective for detecting viral RNA in body fluids and tissues of Marburg- or Ebola-infected patients (Sanchez et al, 1999; Rodriguez et al, 1999). No nucleotide changes were noted in sequence analysis of an RT-PCR fragment of the most variable region of the glycoprotein gene. Ebola virus was detected in a semen specimen up to 91 days following disease onset (Rowe et al, 1999). Filoviruses can persist in tissues and fluids of convalescent patients.
    6) Geisbert & Jaax (1998) described an immunohistochemistry (IHC) and electron and immunoelectron microscopy technique to identify Marburg virus from tissue and fluids isolated from a fatal human case of Marburg hemorrhagic disease (Geisbert & Jaax, 1998). Zaki et al (1999) demonstrated the effectiveness, sensitivity and safety of IHC testing of formalin-fixed skin specimens for laboratory diagnosis of Ebola virus infection (Zaki et al, 1999). This technique has been shown to be effective only in later stages of disease in animals, and not early in the incubation phase (Rollin et al, 1999).
    C) OTHER
    1) Diagnosis of Ebola virus in suspected patients may be accomplished by detection of Ebola antigen in formalin-preserved skin snips (Breman et al, 1997).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.4) DISPOSITION/EYE EXPOSURE
    6.3.4.3) CONSULT CRITERIA/EYE
    A) Persons with conjunctiva exposure to blood, body fluids, secretions, or excretions from a patient with suspected filovirus infection should receive medical evaluation and follow-up management (CDC, 1995).
    6.3.5) DISPOSITION/DERMAL EXPOSURE
    6.3.5.1) ADMISSION CRITERIA/DERMAL
    A) All patients with suspected Ebola or Marburg infections should be admitted to an intensive care unit and strict isolation and barrier nursing techniques should be instituted. Since filoviruses are classified as biosafety level 4 agents, diagnostic work with these agents is limited to a few selected laboratories, such as the CDC in Atlanta, Georgia (Feldman et al, 1996).
    B) Any person who has had close physical contact with patients should be put under strict surveillance (twice daily body temperature checks; in case of temperature >38.3 degrees C (101 degrees F), hospitalize immediately in strict isolation). Hospital personnel who come into close contact with patients or contaminated materials without barrier nursing attire must be considered exposed and put under close, supervised surveillance (WHO, 1995).
    6.3.5.2) HOME CRITERIA/DERMAL
    A) Casual contacts with filovirus patients should be placed on alert and asked to report any fever. All surveillance should be continued for 3 weeks after the date of the last contact (WHO, 1995).
    6.3.5.3) CONSULT CRITERIA/DERMAL
    A) All confirmed and highly probable cases MUST be reported to local or state public health departments. Infectious disease consultation is recommended.

Monitoring

    A) Obtain CBC, electrolytes, PT and PTT, renal function tests, liver enzymes, blood cultures, CSF, ascites fluid, or pleural fluid as clinically indicated. Because filoviruses are classified as biosafety level 4 agents, work with these agents is limited to a few selected laboratories, such as the CDC in Atlanta, Georgia.
    B) Enzyme-linked immunosorbent assay (ELISA) and electrophoretic immunotransblot test (EITB, Western blot) may be valuable serologic diagnostic tests when performed in selected laboratories.
    C) Ebola virus is usually detectable in blood by real-time polymerase chain reaction testing 3 to 10 days post-symptom onset.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) DECONTAMINATION
    1) ORAL: Oral decontamination measures following exposure to Ebola or Marburg viruses have not been proven to be effective and are NOT recommended.
    2) DERMAL: Remove contaminated clothing immediately. Sites exposed to blood/body fluids should be immediately washed with soap and water.
    3) EYE: Remove contact lenses and irrigate exposed eyes with copious amounts of room temperature 0.9% saline or water for at least 15 minutes.
    4) Any person who has had close physical contact with patients should be put under strict surveillance (twice daily body temperature checks; in case of temperature >38.3 degrees C (101 degrees F), hospitalize immediately in strict isolation). Hospital personnel who come into close contact with patients or contaminated materials without barrier nursing attire must be considered exposed and put under close, supervised surveillance (WHO, 1995).
    6.5.2) PREVENTION OF ABSORPTION
    A) Oral decontamination measures following exposure to Ebola or Marburg viruses have not been proven to be effective and are NOT recommended.
    6.5.3) TREATMENT
    A) SUPPORT
    1) Treatment should include recommendations listed in the DERMAL EXPOSURE section when appropriate.

Inhalation Exposure

    6.7.2) TREATMENT
    A) SUPPORT
    1) Although filoviruses are not known to be transmitted from human-to-human via inhalation, researchers have shown a potential of aerogenic infection by Ebola virus administered into the respiratory tract of rhesus monkeys via inhalation (Johnson et al, 1995). Use of Ebola virus as a biological warfare agent has been proposed due to its highly contagious and lethal properties. However, because of uncertain stability outside of animal hosts, it may not be desirable as a biological agent for warfare unless a stable aerosolized form is developed (Cole, 1996). Medical personnel working with known filovirus infected patients should wear protective masks and utilize strict barrier nursing techniques. Respirators should be used when caring for patients with coughing, vomiting, diarrhea or hemorrhage (CDC, 1995c).
    2) Treatment should include recommendations listed in the DERMAL EXPOSURE section when appropriate.
    B) Treatment should include recommendations listed in the ORAL EXPOSURE section when appropriate.

Eye Exposure

    6.8.1) DECONTAMINATION
    A) EYE IRRIGATION, ROUTINE: Remove contact lenses and irrigate exposed eyes with copious amounts of room temperature 0.9% saline or water for at least 15 minutes. If irritation, pain, swelling, lacrimation, or photophobia persist after 15 minutes of irrigation, an ophthalmologic examination should be performed (Peate, 2007; Naradzay & Barish, 2006).

Dermal Exposure

    6.9.1) DECONTAMINATION
    A) DERMAL DECONTAMINATION
    1) DECONTAMINATION: Remove contaminated clothing and wash exposed area thoroughly with soap and water for 10 to 15 minutes. A physician may need to examine the area if irritation or pain persists (Burgess et al, 1999).
    2) Use of an antiseptic solution or hand washing product may be considered, although the efficacy of this supplemental measure is unknown.
    B) CLOTHING
    1) Bag soiled clothing or dressings in clearly labeled leak-proof polyethylene bags until autoclaved or incinerated.
    6.9.2) TREATMENT
    A) SUPPORT
    1) MANAGEMENT OF MILD TO MODERATE TOXICITY
    a) Intensive supportive treatment is the mainstay of therapy. No antidote or vaccine is available for filovirus infections. Correct any significant fluid and/or electrolyte abnormalities in patients with severe diarrhea and/or vomiting.
    2) MANAGEMENT OF SEVERE TOXICITY
    a) Intensive supportive treatment is the mainstay of therapy. No antidote or vaccine is available for filovirus infections. Transfusions of fresh frozen plasma/packed red blood cells, and platelets (as needed) for severe thrombocytopenia, bleeding. Treat severe hypotension with IV 0.9% NaCl at 10 to 20 mL/kg. Add dopamine or norepinephrine if unresponsive to fluids. Hemodialysis may be necessary in cases of renal failure.
    3) Filoviruses may be transmitted in hospital settings (CDC, 1995) (WHO, 1995). Strict BARRIER precautions are required for all patient caregivers. Restrict nonessential staff and visitors from entering the room. Patients should be isolated until they are virus free, or for a 3 week period following illness. During the early stages of infection a negative pressure room is not required, but should be considered at the time of hospitalization to avoid the need for subsequent transfer of the patient. Disinfection of patient's urine, sputum, blood, clothing, and bedding is important. Use disposable syringes and needles.
    a) Contaminated environmental surfaces or inanimate objects should be cleansed and disinfected using standard procedures with EPA registered hospital disinfectant or a 1:100 dilution of household bleach (CDC, 1995).
    4) Since filoviruses are classified as biosafety level 4 agents, diagnostic work with these agents is limited to a few selected laboratories, such as the CDC in Atlanta, Georgia (Feldman et al, 1996). Only specially trained personnel should handle blood and other biologic specimens. Blood and autopsy specimens should be placed in tightly sealed metal containers, wrapped in absorbent material inside a sealed plastic bag, and shipped on dry ice to laboratories with biocontainment level 4 facilities.
    B) FLUID/ELECTROLYTE BALANCE REGULATION
    1) Careful monitoring of electrolytes and proper fluid replacement is essential. (Note: Fluid and electrolyte replacement maybe similar to that seen with cholera patients.) Assess clinical status to determine magnitude of dehydration.
    2) MODERATE TO SEVERE DEHYDRATION (and normal renal function): Rehydrate over 30 to 45 minutes with 1 to 2 liters normal saline or lactated Ringers solution. Repeat a bolus (0.5 to 1 L) over 30 to 45 minutes if response is poor. In children, begin with 10 to 20 milliliters/kilogram of normal saline or lactated Ringers solution.
    3) With rapidly continuing losses, suspected cardiac or renal dysfunction, or inability to stabilize patient, central venous pressure or pulmonary wedge pressure may need to be monitored.
    4) Following stabilization of vital signs and other parameters of severe dehydration, maintenance and deficit fluids can be replaced by a combination of 0.5NS, 0.25NS, and D5W at a rate to maintain normal vital signs and urine output. This assumes that most patients will have isotonic dehydration.
    C) AIRWAY MANAGEMENT
    1) In cases of airway compromise, supportive measures including endotracheal intubation and mechanical ventilation may be necessary.
    D) TRANSFUSION
    1) Administer fresh frozen plasma and packed red blood cells or platelets as needed for active bleeding. Some patients have responded with good results to administration of clotting factor concentrates (Halstead, 1996).
    E) MONITORING OF PATIENT
    1) Obtain CBC, electrolytes, PT and PTT, renal function tests, liver enzyme concentrations, blood cultures, CSF, ascites fluid, or pleural fluid as clinically indicated. Because filoviruses are classified as biosafety level 4 agents, work with these agents is limited to a few selected laboratories, such as the CDC in Atlanta, Georgia.
    2) Enzyme-linked immunosorbent assay (ELISA) and electrophoretic immunotransblot test (EITB, Western blot) may be valuable serologic diagnostic tests when performed in selected laboratories.
    3) Ebola virus is usually detectable in blood by real-time polymerase chain reaction testing 3 to 10 days post-symptom onset.
    F) HYPOTENSIVE EPISODE
    1) SUMMARY
    a) Infuse 10 to 20 milliliters/kilogram of isotonic fluid and keep the patient supine. If hypotension persists, administer dopamine or norepinephrine. Consider central venous pressure monitoring to guide further fluid therapy.
    2) DOPAMINE
    a) DOSE: Begin at 5 micrograms per kilogram per minute progressing in 5 micrograms per kilogram per minute increments as needed (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). If hypotension persists, dopamine may need to be discontinued and a more potent vasoconstrictor (eg, norepinephrine) should be considered (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    b) CAUTION: If ventricular dysrhythmias occur, decrease rate of administration (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). Extravasation may cause local tissue necrosis, administration through a central venous catheter is preferred (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    3) NOREPINEPHRINE
    a) PREPARATION: 4 milligrams (1 amp) added to 1000 milliliters of diluent provides a concentration of 4 micrograms/milliliter of norepinephrine base. Norepinephrine bitartrate should be mixed in dextrose solutions (dextrose 5% in water, dextrose 5% in saline) since dextrose-containing solutions protect against excessive oxidation and subsequent potency loss. Administration in saline alone is not recommended (Prod Info norepinephrine bitartrate injection, 2005).
    b) DOSE
    1) ADULT: Dose range: 0.1 to 0.5 microgram/kilogram/minute (eg, 70 kg adult 7 to 35 mcg/min); titrate to maintain adequate blood pressure (Peberdy et al, 2010).
    2) CHILD: Dose range: 0.1 to 2 micrograms/kilogram/minute; titrate to maintain adequate blood pressure (Kleinman et al, 2010).
    3) CAUTION: Extravasation may cause local tissue ischemia, administration by central venous catheter is advised (Peberdy et al, 2010).
    G) HEMODIALYSIS
    1) Hemodialysis may be necessary in patients with renal failure.
    H) EXPERIMENTAL THERAPY
    1) EBOLA
    a) In the West Africa Ebola outbreak of 2014, the use of convalescent whole blood or plasma collected from patients recovered from Ebola virus disease was evaluated as a matter of immediate priority. Some Ebola treatment centers have administered this empiric treatment (World Health Organization (WHO), 2014).
    1) Donors must be clinically asymptomatic and twice negative for Ebola virus RNA by molecular techniques with samples at least 48 hours apart (World Health Organization, 2014).
    2) Recipients must have confirmed Ebola virus disease, preferably in the early stages (World Health Organization, 2014).
    b) There are several novel therapies being considered to treat Ebola virus disease (World Health Organization (WHO), 2014).
    1) ZMapp(TM) is a combination of 3 humanized monoclonal antibodies that bind to the Ebola Zaire virus strain. The monoclonal antibodies are manufactured from Nicotiana benthamiana (tobacco) plants using recombinant technology. It has demonstrated efficacy in a monkey model of Ebola in studies conducted by the Public Health Agency of Canada. Human data are extremely limited (Mapp Biopharmaceutical, Inc, 2014; Mapp Biopharmaceutical, Inc, 2014).
    2) Brincidofovir has been studied in vitro and in animals and has demonstrated antiviral activities. The US Food and Drug Administration has granted emergency Investigational New Drug Application to brincidofovir to be used in patients with Ebola Virus Disease (Chimerix, 2014).
    3) Favipiravir is a pyrazinecarboxamide derivative antiviral approved in Japan for the treatment of influenza virus infection. It is a selective inhibitor of the influenza virus RNA polymerase. It was effective against Zaire Ebola virus in vitro and in vivo studies and has been administered to Ebola infected human on an individual basis (Fujifilm Corporation, 2014).
    4) BCX4430 is an RNA dependent-RNA polymerase inhibitor has broad-spectrum activity against more than 20 RNA viruses in 9 different families, including filoviruses, togaviruses, bunyaviruses, arenaviruses, paramyxoviruses, coronaviruses and flaviviruses (BioCryst Pharmaceuticals, Inc, 2014).
    5) TKM-Ebola is a small interfering RNA (siRNA) targeting the Ebola virus delivered by the lipid nanoparticle (LNP) technology. It was used to treat non-human primates infected with Zaire Ebola virus with 100% protection against the infection. It is currently in Phase 1 trials (Tekmira Pharmaceuticals Corporation, 2014).
    c) In a case of accidental injection of Ebola virus (from an infected guinea pig) in a laboratory worker, Edmond et al (1977) described treatment with interferon (3 million units IM every 12 hours for 14 days) which was started 20 hours after the onset of the patient's illness. Convalescent serum, from patients recovering from filovirus infection, was given 47 hours after onset of illness, as a 4 hour infusion. On the first and second day of illness, viral concentrations in the blood were at peak levels. Viral concentrations dropped after the start of interferon and serotherapy and remained low until the viremia disappeared on the ninth day of illness. This patient had a relatively mild course of illness and an absence hemorrhage (Edmond et al, 1977). The value of this therapy cannot be accurately assessed without further experiences (Mupapa et al, 1999). Sadek et al (1999) reported no statistical evidence of survival benefit following transfusions of convalescent serum (Sadek et al, 1999).
    d) An immunoglobulin to Ebola virus infection has been formed from hyperimmune equine blood sera. The immunoglobulin has been given to monkeys infected intramuscularly with doses of 110 to 29 LD50 Ebola virus and shown to offer protection up to 100%. The immunoglobulin has not been tested in humans (Borisevich et al, 1995) but has shown some activity in suppressing viremia and delaying disease onset and death in non-human primates.
    e) Wimer (2002) has suggested that intravenous administration of plant mitogens (particularly PHA) may be able to inhibit Ebola virus glycoprotein effects, that cause vascular cellular damage, as well as interacting with T lymphocytes to stimulate the entire immune system, thereby supporting myelopoiesis and lymphopoiesis, in the event of bioweaponry/bioterrorist assaults (Wimer, 2002).
    f) Research and construction of IgG Fab anti-Ebola antibodies is occurring, but is currently in the early phases of development and animal trials (Breman et al, 1997). Non-human primates and guinea pigs will be used to test the immunologic interventions.
    g) Development and animal testing of an anti-Ebola vaccine is being investigated (Breman et al, 1997).
    h) A study was conducted to evaluate the effects of an anti-Ebola vaccine on non-human primates that was previously successful in protecting mice and guinea pigs from an Ebola viral infection. The vaccine strategies that were used included one of four antigens: RNA replicon particles derived from an attenuated strain of Venezuelan equine encephalitis virus expressing the Ebola virus (EBOV) glycoprotein and nucleoprotein, the recombinant Vaccinia virus expressing EBOV glycoprotein, liposomes containing lipid A and inactivated EBOV, or a concentrated and inactivated whole-virion preparation. All of the primates involved in the study (n=26), including 4 primates that were not given one of the vaccine strategies, were challenged with a Zaire subtype of EBOV, which was isolated from a human patient in 1995. Twenty-five of the 26 primates died within 11 days post-challenge. The one primate that did survive the challenge received the inactivated virion antigen and did not become ill during the study. The primate's neutralizing antibody titers were greater than 320 at day 26 post-challenge and then decreased to 80 at days 26, 61, 99, and 902 post-challenge. The results of this study show that the vaccine strategies, which were successful in protecting rodents from EBOV, are not effective in protecting non-human primates from EBOV. It is suggested that this inconsistency in vaccine efficacy may be due to differences in disease pathology between rodents and primates (Geisbert et al, 2002).
    i) A study, involving rhesus macaque monkeys, was conducted to determine the efficacy of a recombinant nematode anticoagulant protein (an inhibitor of factor VIIa/tissue factor) in alleviating effects of Ebola virus infection and subsequently prolonging survival time. The recombinant protein (rNAPc2) was administered to the monkeys either 10 minutes (n=6) or 24 hours (n=3) after a high-dose subcutaneous administration of the Zaire subtype Ebola virus. Three of the monkeys were untreated Ebola virus-positive controls. In the 24-hour group, 1 of the 3 monkeys survived the challenge and remained healthy for at least 1 year. The other two monkeys died within 14 days post-challenge. In the 10-minute group, 2 of the 6 monkeys survived and remained healthy at least 9 months post-challenge. The other four monkeys died within 14 days post-challenge (Geisbert et al, 2003).
    1) With regards to clinical presentation of the infection, the rNAPc2-treated animals developed cutaneous macular rashes much more slowly than the untreated controls. The rashes in the treated monkeys were only observed just before death, as compared with the untreated controls who developed the rashes several days before death.
    2) To determine the recombinant protein effects on the development of coagulopathy during Ebola virus infection, D-dimer was measured in all treated and untreated monkeys. Increased plasma concentrations of D-dimer, indicating an increased risk for disseminated intravascular coagulation, was noted in all of the untreated monkeys but only in 2 of the treated monkeys six days post-challenge. Also, increased plasma concentrations of the inflammatory mediators, interleukin-6 and monocyte chemoattractant protein-1 (MCP-1) , were detected in 3 and 4 of the treated monkeys, respectively, as compared with all of the untreated controls, indicating that the rNAPc2 may attenuate the pro-inflammatory response observed with Ebola virus infections.
    I) INFECTIOUS DISEASE NOTIFICATION
    1) REPORTING/LABORATORY TESTING: All confirmed and suspected cases should be reported immediately through local and state health departments to the Viral Special Pathogens Branch, Centers for Disease Control (CDC), Atlanta, GA. The CDC has a Biosafety Level 4 laboratories, but NO specimen can be accepted without prior consultation. Contact: 404-639-1115 (website: http://www.cdc.gov/ncezid/dhcpp/vspb).
    J) Treatment should include recommendations listed in the ORAL EXPOSURE section when appropriate.

Enhanced Elimination

    A) HEMODIALYSIS
    1) Hemodialysis is unlikely to be necessary, but may be needed in patients that develop acute renal failure.

Case Reports

    A) ACUTE EFFECTS
    1) EBOLA VIRUS
    a) A laboratory worker accidentally injected his finger while transferring homogenized liver from a guinea pig infected with Ebola virus. Following immersion of his finger in a hypochlorite solution, he squeezed it, with no blood appearing. Six days later he became ill with fever, nausea, and abdominal pain. He was transferred to a high-security infectious disease hospital unit and placed in a Trexler negative-pressure plastic isolator. He complained of exhaustion, anorexia, nausea, and abdominal pain. No other symptoms were apparent. Temperature of 38 degrees C was recorded, as well as relative bradycardia.
    1) Treatment with interferon (3 million units IM every 12 hours for 14 days) was started 20 hours after the onset of illness. Convalescent serum was given 47 hours after onset of illness. Between the fourth and seventh day of his illness, symptoms were at their height. Severe weakness, profuse watery diarrhea, persistent vomiting, albuminuria, and a confluent rash covering most of his body developed. Sore throat, with Candidiasis, occurred and was treated. His urinary output dropped to its lowest level, despite adequate fluid replacement. His general condition began to improve after approximately 10 days of illness, with fever subsiding and rash improving. A slow recovery ensued over a 10 week period.
    2) On the first and second day of illness, viral concentrations in the blood were at peak levels. Viral concentrations dropped after the start of interferon and serotherapy and remained low until the viremia disappeared on the ninth day of illness (Edmond et al, 1977).
    2) MARBURG VIRUS
    a) A 15-year-old boy, in Kenya for one month, was admitted to the hospital with a 3 day history of fever, headache, malaise, anorexia, and vomiting. During his hospital course, he developed bloody diarrhea, hypotension, leukocytosis, thrombocytopenia, and disseminated intravascular coagulation (DIC). Supportive therapy included antibiotics, steroids, heparin, fresh plasma, and blood transfusions. The patient died on the eleventh hospital day (Geisbert & Jaax, 1998).
    1) Tissue samples sent to the United States Army Medical Research Institute of Infectious Diseases (USAMRIID) were evaluated and found to be positive for Marburg virus. Necropsy examination showed extensive petechial and purpuric hemorrhage in the skin, conjunctiva, and gastrointestinal mucosa. Hemorrhage of the lungs and tracheobronchial tree were noted. Pleural, pericardial, and peritoneal effusions were bloody. The epicardium, renal cortex and pelvis, and urinary bladder were observed to have multifocal petechial hemorrhages. No hemorrhage was noted in the liver, spleen, pancreas, or adrenals.

Summary

    A) TOXICITY: Animal experiments have shown fatal inhalation doses of Ebola virus to be 400 plaque forming units of virus.

Minimum Lethal Exposure

    A) ANIMAL DATA
    1) INHALATION: Experiments in rhesus monkeys revealed inhalation of viral doses as low as 400 plaque forming units of Ebola virus caused a rapidly fatal disease in 4 to 5 days (Johnson et al, 1995; Tomera, 1997).
    2) INOCULATION: McCormick et al (1983) reported that fewer than 10 infectious particles of a Zaire Ebola strain were lethal for suckling mice, whereas 10,000 infectious particles of a Sudan Ebola strain were unable to kill any of the mice. LD50 of the Zaire strain was calculated to be approximately one infectious particle (McCormick et al, 1983).
    a) Bray et al (1999) reported that the Zaire Ebola virus strain is lethal to suckling mice, but adult mice appear to be resistant to this strain. It was reported that the LD50 in suckling mice was approximately 1 virion (Bray et al, 1999).

Serum Plasma Blood Concentrations

    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) ACUTE
    a) The highest Ebola viral concentrations have been reported in the blood, with lower levels present in throat washings and urine (WHO, 1978a; WHO, 1978b; Johnson et al, 1995).

Toxicologic Mechanism

    A) Filoviruses replicate in endothelial cytoplasm causing focal necrosis, separation of tight junctions and basement membrane detachment (Tomera, 1997). Following viral replication in many organs, focal necrosis has been reported to occur in the liver, spleen, kidneys, lungs, testis and lymphatics, with widespread vascular damage due to impaired microcirculation (Borio et al, 2002). Significant injury to the microvasculature of all organs occurs. Replication of the virus in lymphocytes and monocytes and in interstitial cells of the testis has also been reported. The blood fails to clot, and main lesions appear in the vascular endothelium and the platelets, resulting in severe bleeding in the liver, mucosa, abdomen, pericardium, and other organs. DIC and thrombocytopenia are common and correlate with severity of disease. High fever persists.
    B) Partial renal cellular injury is caused by direct virus invasion of glomerular endothelium and tubular epithelium. Ischemia resulting from thrombosis in the peritubular capillaries is probably the cause of much of the renal tubular damage (Tomera, 1997).
    C) Macrophages and monocytes are reported as primary cellular targets for both Ebola and Marburg virus infection. Viral replication sites included hepatocytes, adrenal cortical and medullary cells, fibroblast-like cells, and endothelial cells (Wyers et al, 1999; Ryabchikova et al, 1999; Connolly et al, 1999; Geisbert & Jaax, 1998).
    D) In chimpanzee studies, Wyers et al (1999) reported the predominant Ebola viral lesions to consist of multifocal necrosis in the liver and diffuse fibrinoid necrosis in the red pulp of the spleen. Macrophages within these sites were shown to contain large eosinophilic intracytoplasmic inclusion bodies. An absence of bronchiolar and pulmonary lesions was noted, suggesting that aerosol transmission was unlikely.
    E) Impairment of immune response to filovirus infection may occur as a result of the secretory glycoprotein (SGP) of Ebola virus inhibiting early activation of neutrophils. Binding of the transmembrane glycoprotein (GP) of the Ebola virus to endothelial cells may contribute to the hemorrhagic manifestations of the disease and disseminated intravascular coagulation (Borio et al, 2002; Takada & Kawaoka, 1998). Volchkov et al (1998) speculated that proteolytic processing of GP could determine the pathogenicity of Ebola virus. The Reston subtype has low human pathogenicity, and has a reduced cleavability due to mutation at the cleavage site.
    F) Marburg-induced hemorrhagic diathesis and shock may be due to viral replication of the Marburg virus in endothelial cells and their subsequent destruction (Borio et al, 2002).
    G) Ebola antibody in infected patients is not always detected prior to death. However, IgG antibody has been detected up to 2 years following onset of the disease in convalescent patients, and up to 10 years in two patients. It has been shown that IgG and IgM antibody appear at about the same time after disease onset (8-10 days), but IgM persists for a much shorter period of time in convalescent patients (Ksiazek et al, 1999; Ksiazek et al, 1999a).
    H) It is strongly suspected that bats, especially solitary microchiropteran species, act as reservoir hosts for Ebola viruses (Monath, 1999).

General Bibliography

    1) Al-Ahdal M: Ebola virus: an update. Saudi Pharm J 1995; 3:135-136.
    2) Anon: Ebola haemorrhagic fever. Weekly Epidemiology Record 1995; 70(20):147-148.
    3) Anon: Outbreak of Ebola haemorrhagic fever in Gabon. Commun Dis Rep CDR Wkly 1996; 6(9):75-78.
    4) Baron RC, McCormick JB, & Zubeir OA: Ebola virus disease in southern Sudan: hospital dissemination and intrafamilial spread. Bull World Health Org 1983; 61:997-1003.
    5) BioCryst Pharmaceuticals, Inc: BioCryst receives additional NIAID funding for manufacture and development of BCX4430 to treat hemorrhagic virus diseases. BioCryst Pharmaceuticals, Inc. Research Triangle Park, NC. 2014. Available from URL: http://files.shareholder.com/downloads/BCRX/3532990615x0x782407/39c9537e-2907-4069-aaae-d507f1e19649/BCRX_News_2014_9_18_General_News.pdf. As accessed 2014-10-09.
    6) Borio L, Inglesby T, & Peters CJ: Hemorrhagic fever viruses as biological weapons - medical and publich health management (Consensus statement). JAMA 2002; 287:2391-2405.
    7) Borisevich IV, Mikhailov VV, & Krasnyansky VP: Creation and study of immunoglobulin to Ebola fever. Vaprosy Virusologii 1995; 40(6):270-273.
    8) Bray M, Davis K, & Geisbert T: A mouse model for evaluation of prophylaxis and therapy of Ebola hemorrhagic fever. J Infect Dis 1999; 179 (Suppl 1):S248-258.
    9) Breman JG, van der Groen G, & Peters CJ: International colloquium on Ebola virus research: summary report. J Infect Dis 1997; 176:1058-1063.
    10) Burgess JL, Kirk M, Borron SW, et al: Emergency department hazardous materials protocol for contaminated patients. Ann Emerg Med 1999; 34(2):205-212.
    11) Butler JC, Kilmarx PH, & Jernigan DB: Perspectives in fatal epidemics. Infect Dis Clinics North America 1996; 10:918-927.
    12) Bwaka MA, Bonnet MJ, & Calain P: Ebola hemorrhagic fever in Kikwit, Democratic Republic of the Congo: clinical observations in 103 patients. J Infect Dis 1999; 179 (Suppl 1):S1-7.
    13) CDC: Ebola virus infection in imported primates - Virginia, 1989. CDC: MMWR 1989; 38:830-838.
    14) CDC: Management of patients with suspected viral hemorrhagic fever. CDC: MMWR 1988; 37:1-16.
    15) CDC: Outbreak of Ebola viral hemorrhagic fever - Zaire, 1995. CDC: MMWR 1995a; 44:381-382.
    16) CDC: Update: Filovirus infection in animal handlers. CDC: MMWR 1990; 39:221.
    17) CDC: Update: Management of patients with suspected viral hemorrhagic fever - United States. CDC: MMWR 1995c; 44:475-479.
    18) CDC: Update: Outbreak of Ebola viral hemorrhagic fever - Zaire, 1995. CDC: MMWR 1995b; 44:469-475.
    19) Centers for Disease Control and Prevention (CDC): 2014 Ebola Outbreak in West Africa - Outbreak Distribution Map. Centers for Disease Control and Prevention (CDC). Atlanta, GA. 2014. Available from URL: http://www.cdc.gov/vhf/ebola/outbreaks/2014-west-africa/distribution-map.html. As accessed 2014-10-06.
    20) Centers for Disease Control and Prevention (CDC): About Ebola Virus Disease. Centers for Disease Control and Prevention (CDC). Atlanta, GA. 2014a. Available from URL: http://www.cdc.gov/vhf/ebola/about.html. As accessed 2014-10-06.
    21) Centers for Disease Control and Prevention (CDC): Ebola Virus Disease: Signs and Symptoms. Centers for Disease Control and Prevention (CDC). Atlanta, GA. 2014f. Available from URL: http://www.cdc.gov/vhf/ebola/symptoms/index.html. As accessed 2014-10-06.
    22) Centers for Disease Control and Prevention (CDC): Ebola Virus Disease: Transmission. Centers for Disease Control and Prevention (CDC). Atlanta, GA. 2014d. Available from URL: http://www.cdc.gov/vhf/ebola/transmission/index.html. As accessed 2014-10-06.
    23) Centers for Disease Control and Prevention (CDC): Interim Guidance for Specimen Collection, Transport, Testing, and Submission for Persons Under Investigation for Ebola Virus Disease in the United States. Centers for Disease Control and Prevention (CDC). Atlanta, GA. 2014b. Available from URL: http://www.cdc.gov/vhf/ebola/hcp/interim-guidance-specimen-collection-submission-patients-suspected-infection-ebola.html. As accessed 2014-10-08.
    24) Centers for Disease Control and Prevention (CDC): Marburg hemorrhagic fever (Marburg HF). Centers for Disease Control and Prevention (CDC). Atlanta, GA. 2014c. Available from URL: http://www.cdc.gov/vhf/marburg/index.html. As accessed 2015-01-27.
    25) Centers for Disease Control and Prevention (CDC): Marburg hemorrhagic fever (Marburg HF): Transmission. Centers for Disease Control and Prevention (CDC). Atlanta, GA. 2014e. Available from URL: http://www.cdc.gov/vhf/marburg/transmission/index.html. As accessed 2015-01-27.
    26) Chimerix INC: Frequently asked questions on the potential use of brincidofovir in the Ebola outbreak. Chimerix, Inc. Durham, NC. 2014. Available from URL: http://www.chimerix.com/c/discovery-clinical-trials/brincidofovir-ebola.php. As accessed 2014-10-09.
    27) Cole LA: The specter of biological weapons. Sci Amer 1996; 275:60-65.
    28) Connolly BM, Steele KE, & Davis KJ: Pathogenesis of experimental Ebola virus infection in guinea pigs. J Infect Dis 1999; 179(Suppl 1):S203-217.
    29) Dalgard DW, Hardy RJ, & Pearson SL: Combined Simian hemorrhagic fever and Ebola virus infection in cynomolgus monkeys. Lab Animal Sci 1992; 42:152-157.
    30) Dowell SF, Mukunu R, & Ksiazek TG: Transmission of Ebola hemorrhagic fever: a study of risk factors in family members, Kikwit, Democratic Republic of the Congo, 1995. J Infect Dis 1999; 179(Suppl 1):S87-91.
    31) Edmond RTD, Evans B, & Bowen ETW: A case of Ebola virus infection. Br Med J 1977; 2:541-544.
    32) Feldman H, Slenczka W, & Klenk HD: Emerging and reemerging of filoviruses. Arch Virol 1996; 11 (Suppl):77-100.
    33) Formenty P, Hatz C, & Le Guenno B: Human infection due to Ebola virus, subtype Cote d' Ivoire: clinical and biologic presentation. J Infect Dis 1999; 179(Suppl 1):S48-53.
    34) Fujifilm Corporation: Avigan(R) Tablet 200mg administered to a French woman infected with Ebola virus. Fujifilm Corporation. Tokyo, Japan. 2014. Available from URL: http://www.fujifilm.com/news/n140926.html. As accessed 2014-10-09.
    35) Geisbert T, Pushko P, Anderson K, et al: Evaluation in nonhuman primates of vaccines against Ebola virus. Emerging Infect Dis 2002; 8:503-507.
    36) Geisbert TW & Jaax NK: Marburg hemorrhagic fever: report of a case studied by immunohistochemistry and electron microscopy. Ultrastructural Pathology 1998; 22:3-17.
    37) Geisbert TW, Hensley LE, Jahrling PB, et al: Treatment of Ebola virus infection with a recombinant inhibitor of factor VIIa/tissue factor: a study in rhesus monkeys. Lancet 2003; 362:1953-1958.
    38) Halstead S: Arboviruses, in Behrman (ed): Nelson Textbook of Pediatrics, 15th ed, WB Saunders, Philadelphia, PA, 1996.
    39) Hayes CG, Burans JP, & Ksiazek TG: Outbreak of fatal illness among captive macaques in the Philippines caused by an Ebola-related filovirus. Am J Trop Med Hyg 1992; 46:664-671.
    40) Jaax NK, Davis KJ, & Geisbert TJ: Lethal experimental infection of rhesus monkeys with Ebola-Zaire (Mayinga) virus by the oral and conjunctival route of exposure. Arch Pathol Lab Med 1996; 120:140.
    41) Jahrling PB, Geisbert TW, & Dalgard DW: Preliminary report: isolation of Ebola virus from monkeys imported to USA. Lancet 1990; 335:502-505.
    42) Johnson E, Jaax N, & White J: Lethal experimental infections of rhesus monkeys by aerosolized Ebola virus. Int J Exp Path 1995; 76:227-236.
    43) Johnson ED, Johnson BK, & Silverstein D: Characterization of a new Marburg virus isolated from a 1987 fatal case in Kenya. Arch Virol 1996; 11(Suppl):101-114.
    44) Kalongi Y, Mwanza K, & Tshisuaka M: Isolated case of Ebola hemorrhagic fever with mucormycosis complications, Kinshasa, Democratic Republic of the Congo. J Infect Dis 1999; 179(Suppl 1):S15-17.
    45) Kalter SS, Heberling RL, & Barry JD: Detection of Ebola-Reston (filoviridae) virus antibody by Dot-immunobinding assay. Lab Animal Sci 1995; 45:523-525.
    46) Kibadi K, Mupapa K, & Kuvula K: Late ophthalmologic manifestations in survivors of the 1995 Ebola virus epidemic in Kikwit, Democratic Republic of the Congo. J Infect Dis 1999; 179(Suppl 1):S13-14.
    47) Kleinman ME, Chameides L, Schexnayder SM, et al: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Part 14: pediatric advanced life support. Circulation 2010; 122(18 Suppl.3):S876-S908.
    48) Ksiazek TG, Rollin PE, & Williams AJ: Clinical virology of Ebola hemorrhagic fever (EHF): virus, virus antigen, and IgG and IgM antibody finding among EHF patients in Kikwit, Democratic Republic of the Congo, 1995. J Infect Dis 1999; 179 (Suppl 1):S177-187.
    49) Ksiazek TG, West CP, & Rollin PE: ELISA for the detection of antibodies to Ebola viruses. J Infect Dis 1999a; 179(Suppl 1):S192-198.
    50) Le Guenno B, Formentry P, & Wyers M: Isolation and partial characterisation of a new strain of Ebola virus. Lancet 1995; 345:1271-1274.
    51) Leroy EM, Baize S, & Volchkov VE: Human asymptomatic Ebola infection and strong inflammatory response. Lancet 2000; 355:2210-2215.
    52) Leroy EM, Souquiere S, Rouquet P, et al: Re-emergence of ebola haemorrhagic fever in Gabon (letter). Lancet 2002; 359:712.
    53) Lub M, Sergeev AN, & P'iankova OG: Clinical-virusological characteristics of disease in guinea pigs, infected by the Marburg virus aerogenically (abstract). Voprosy Virusologii 1995; 40(3):119-121.
    54) Lupton HW: Inactivation of Ebola virus with 60Co irradiation. J Infect Dis 1981; 143:291.
    55) Mapp Biopharmaceutical, Inc: ZMapp(TM) frequently asked questions. Mapp Biopharmaceutical, Inc. San Diego, CA. 2014. Available from URL: http://www.mappbio.com/zmappfaq.pdf. As accessed 2014-10-09.
    56) Mapp Biopharmaceutical, Inc: ZMapp(TM) information sheet. Mapp Biopharmaceutical, Inc. San Diego, CA. 2014a. Available from URL: http://www.mappbio.com/zmapinfo.pdf. As accessed 2014-10-09.
    57) McCormick JB, Bauer SP, & Elliott LH: Biologic differences between strains of Ebola virus from Zaire and Sudan. J Infect Dis 1983; 147:264-267.
    58) Merzlikin NV, Chepurnov AA, & Istomina NN: Development and use of enzyme immunoassay test systems for the diagnosis of Ebola fever. Vop Virusol 1995; 40:31-35.
    59) Monath TP: Ecology of Marburg and Ebola viruses: speculations and directions for future research. J Infect Dis 1999; 179(Suppl 1):S127-138.
    60) Mupapa K, Massamba M, & Kibadi K: Treatment of Ebola hemorrhagic fever with blood transfusions from convalescent patients. J Infect Dis 1999; 179(Suppl 1):S18-23.
    61) Naradzay J & Barish RA: Approach to ophthalmologic emergencies. Med Clin North Am 2006; 90(2):305-328.
    62) Peate WF: Work-related eye injuries and illnesses. Am Fam Physician 2007; 75(7):1017-1022.
    63) Peberdy MA , Callaway CW , Neumar RW , et al: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care science. Part 9: post–cardiac arrest care. Circulation 2010; 122(18 Suppl 3):S768-S786.
    64) Peters CJ: Emerging infections: Ebola and other filoviruses. West J Med 1996; 164:36-38.
    65) Product Information: dopamine hcl, 5% dextrose IV injection, dopamine hcl, 5% dextrose IV injection. Hospira,Inc, Lake Forest, IL, 2004.
    66) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    67) Richards GA, Murphy S, & Jobson R: Unexpected Ebola virus in a tertiary setting: clinical and epidemiologic aspects. Crit Care Med 2000; 28:240-244.
    68) Rodriguez LL, De Roo A, & Guimard Y: Persistence and genetic stability of Ebola virus during the outbreak in Kikwit, Democratic Republic of the Congo, 1995. J Infect Dis 1999; 179(Suppl 1):S170-176.
    69) Roels TH, Bloom AS, & Buffington J: Ebola hemorrhagic fever, Kikwit, Democratic Republic of the Congo, 1995; risk factors for patients without a reported exposure. J Infect Dis 1999; 179(Suppl 1):S92-97.
    70) Rollin PE & Ksiazek TG: Ebola haemorrhagic fever. Trans Roy Soc Trop Med Hyg 1998; 92:1-2.
    71) Rollin PE, Williams RJ, & Bressler DS: Ebola (subtype Reston) virus among quarantined nonhuman primates recently imported from the Philippines to the United States. J Infect Dis 1999; 179 (Suppl):S108-S114.
    72) Rowe AK, Bertolli J, & Khan AS: Clinical, virologic, and immunologic follow-up of convalescent Ebola hemorrhagic fever patients and their household contacts, Kikwit, Democratic Republic of the Congo. J Infect Dis 1999; 179(Suppl 1):S28-35.
    73) Ryabchikova EI, Kolesnikova LV, & Luchko SV: An analysis of features of pathogenesis in two animal models of Ebola virus infection. J Infect Dis 1999; 179(Suppl 1):S199-202.
    74) Sadek RF, Khan AS, & Stevens G: Ebola hemorrhagic fever, Democratic Republic of the Congo, 1995: determinants of survival. J Infect Dis 1999; 179(Suppl 1):S24-27.
    75) Sanchez A, Ksiazek TG, & Rollin PE: Detection and molecular characterization of Ebola viruses causing disease in human and nonhuman primates. J Infect Dis 1999; 179(Suppl 1):S164-169.
    76) Shope RE: Introduction to hemorrhagic fever viruses, in Bennett(ed): Textbook of Medicine, 20th ed, WB Saunders, Philadelphia, PA, 1996.
    77) Sodhi A: Ebola virus disease; recognizing the face of a rare killer. Postgrad Med 1996; 99:75-78.
    78) Takada A & Kawaoka Y: Pathogenesis of Ebola virus infection: recent insights. Trends in Microbiology 1998; 6:258-259.
    79) Tekmira Pharmaceuticals Corporation: Tekmira doses first subject in human clinical trial of TKM-Ebola. Tekmira Pharmaceuticals Corporation. Burnaby, BC, Canada. 2014. Available from URL: http://investor.tekmirapharm.com/releasedetail.cfm?ReleaseID=819313. As accessed 2014-10-09.
    80) Tomera JF: Threat of Ebola viral hemorrhagic fevers: pharmacologic prospects for a recently emerged member of the filovirus family. Drugs of Today 1997; 33:51-57.
    81) Villinger F, Rollin PE, & Brar SS: Markedly elevated levels of interferon (IFN)-y, IFN-a, interleukin (IL)-2, IL-10, and tumor necrosis factor-a associated with fatal Ebola virus infection. J Infect Dis 1999; 179 (Suppl 1):S188-S191.
    82) WHO: World Health Organization: Ebola haemorrhagic fever in Sudan. WHO: Bull WHO 1978a; 56:247-270.
    83) WHO: World Health Organization: Ebola haemorrhagic fever in Zaire, 1976. WHO: Bull WHO 1978b; 56:271-293.
    84) WHO: World Health Organization: Ebola haemorrhagic fever. WHO: Weekly Epidemiology Record 1995; 70(21):149-151.
    85) Wimer BM: Mitogen therapy for biological warfare/terrorist attacks and viral hemorrhagic fever control. Canc Biother Radiopharmaceut 2002; 17:19-28.
    86) World Health Organization (WHO): Ebola treatment and interventions. World Health Organization (WHO). Geneva, Switzerland. 2014. Available from URL: http://www.who.int/medicines/emp_ebola_q_as/en/. As accessed 2014-10-08.
    87) World Health Organization: Use of convalescent whole blood or plasma collected from patients recovered from Ebola virus disease for transfusion, as an empirical treatment during outbreaks. World Health Organization. Geneva, Switzerland. 2014. Available from URL: http://apps.who.int/iris/bitstream/10665/135591/1/WHO_HIS_SDS_2014.8_eng.pdf?ua=1. As accessed 2014-10-06.
    88) Wyers M, Formenty P, & Cherel Y: Histopathological and immunohistochemical studies of lesions associated with Ebola virus in a naturally infected chimpanzee. J Infect Dis 1999; 179(Suppl 1):S54-59.
    89) Zaki SR, Shieh WJ, Greer PW, et al: A novel immunohistochemical assay for the detection of Ebola virus in skin: implications for diagnosis, spread, and surveillance of Ebola hemorrhagic fever. Commission de Lutte contre les Epidemies a Kikwit. J Infect Dis 1999; 179 Suppl 1:S36-S47.