The liver is a major blood-filtering organ and consequently is predisposed to blood-borne infections causing infectious hepatitis. Viral infections tend to affect the liver in particular, and because of their high affinity for the liver, a subgroup of viruses is termed hepatitis viruses. This group is covered in separate chapters of this book. Here, we summarize other viral infections that do not primarily target the liver but can cause viral hepatitis as part of a systemic infection or lead to hepatic complications under certain conditions, such as immunodeficiency. Of course, this chapter cannot replace a clinical textbook on viral infections but instead focuses on the hepatic aspects of viral infections.
Infections caused by viruses other than the hepatitis viruses can roughly be classified into three major categories: liver disease in patients with fever returning from tropical and subtropical areas, which is frequently caused by exotic agents; severe liver damage in patients with immunodeficiency as a result of de novo infection or exacerbations of common agents such as herpesviruses or adenoviruses; and liver involvement in patients with respiratory and systemic infections, frequently mediated via immunologic mechanisms.
Viral Hemorrhagic Fevers
Approximately 8% of travelers to the developing world require medical care during or after travel, and fever is the underlying problem in 28% of them.1, 2 Physicians evaluating returned travelers with fever frequently suspect rare or exotic diagnoses. Travel-associated liver disease caused by exotic infections such as Ebola virus, Rift Valley fever, or Lassa fever has been reported sporadically in the literature but currently does not represent a frequent health problem. Of the identified causes, dengue is among the top three etiologic agents, and it accounts for approximately 6% of febrile illnesses in travelers.2 Of note, although malaria is the leading cause of systemic febrile illness worldwide, travelers from every tropical or subtropical region except sub-Saharan Africa and Central America have confirmed or suspected dengue more frequently than malaria. Chikungunya is an emerging novel viral infection that has recently been reported in Asia and Africa to cause fever with prominent myalgia, arthralgia, and rash in increasing numbers of patients.3
Viral hemorrhagic fevers share some epidemiologic and clinical features and cause rather similar liver pathology. Most viruses are transmitted via arthropod vectors. The various viruses cause damage to small vessels in multiple organs, which frequently leads to overt hemorrhage. The spectrum of diseases and their geographic distribution are listed in Table 34-1 . Much attention has been paid to abnormal liver function and altered hepatic pathology. Nevertheless, clinically significant liver disease and death from liver failure are rare complications except in patients with yellow fever.
Table 34-1.
Viral Hemorrhagic Fevers Affecting the Liver
DISEASE | VIRUS GROUP | GEOGRAPHIC REGION |
---|---|---|
Dengue | Flaviviridae | Africa, Asia, tropical America |
Yellow fever | Flaviviridae | Africa, South America |
Lassa fever | Arenaviridae | West Africa |
Argentine hemorrhagic fever (Junin virus) | Arenaviridae | Argentina |
Ebola fever | Filoviridae | Central and western Africa |
Marburg fever | Filoviridae | Central and southern Africa |
Rift Valley fever | Bunyaviridae | East and central Africa |
Congo-Crimea hemorrhagic fever | Bunyaviridae | Former Soviet Union, central-western Asia, Africa |
Hemorrhagic fever with renal syndrome | Bunyaviridae | Northern Eurasia |
Chikungunya | Togaviridae | West Africa, Asia, Oceania, southern Europe |
Dengue Fever
The dengue virus complex consists of four antigenically related but distinct flaviviruses termed dengue virus serotypes 1 through 4 (DEN1 to DEN4).4 Dengue viruses are transmitted by Aedes aegypti mosquitos in epidemic and endemic outbreaks and cause acute infections. Three to 6 days after a mosquito bite the virus spreads via the bloodstream, and among the various organs it can be isolated frequently from liver samples,5 where dengue viral antigens have been detected in Kupffer cells, sinusoidal endothelial cells, and hepatocytes.6, 7 Dengue virus infection usually causes a flulike illness with a rash—dengue fever. Hepatomegaly and elevated serum aminotransferases, which are usually mild, are common in dengue virus infections.8, 9 Clinically more severe diseases, such as dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS), can follow secondary infection with dengue virus of a different serotype, thus suggesting that immune responses generated during previous exposure to dengue viruses may enhance the damage triggered by secondary dengue infection. In DHF there are widespread petechial hemorrhages together with multiple organ damage; in DSS, which mostly affects children younger than 15 years, there is extensive capillary leakage and severe fluid depletion leading to hypovolemic shock. If untreated, mortality approaches 50%. In fatal cases of DHF the liver is enlarged, is pale as a result of steatosis, and shows multifocal hemorrhages. Microscopically, focal hepatocellular necrotic areas are seen, as well as coalescent perivenular and midzonal necrosis with Councilman bodies, but relatively little inflammatory cell infiltration.10, 11 It is supposed that liver injury is mediated by dengue virus infection of hepatocytes and Kupffer cells, as in vitro infection of a human hepatoma cell line with dengue virus has been shown to induce apoptotic cell death.12
Currently, a vaccine against dengue virus is not available, and treatment is supportive, with DSS requiring intensive care.
Yellow Fever
Yellow fever is the prototype member of the Flaviviridae family, a group of plus-strand, single-stranded RNA viruses. The yellow fever virus has a single conserved serotype and seven major genotypes reflecting distinct regions in western Africa, central-east Africa, and South America.13, 14 Yellow fever virus is transmitted by a variety of different Aedes vectors and causes endemic and epidemic outbreaks in Africa and South America. Approximately 200,000 cases of yellow fever are currently still reported per year, with 90% occurring in Africa. Yellow fever in travelers to Africa and South America has become rare since the introduction of routine vaccination.
The spectrum of infection with yellow fever virus ranges from subclinical infection to a life-threatening disease with fever, jaundice, renal failure, and hemorrhage. Usually, yellow fever initially appears as an acute, flulike illness of sudden onset with fever, myalgia, and headache that cannot easily be distinguished from other acute infections.15 Between 48 and 72 hours after onset, serum aminotransferases start to rise, thus heralding the development of jaundice. The degree of liver abnormalities at this stage predicts the severity of liver disease later during the course of the illness.16 Next, a period of remission lasting up to 48 hours may follow the initial infection. Patients with abortive infection recover at this stage, but approximately 15% of patients will enter the third stage of intoxication, which is characterized by the return of fever, prostration, and organ dysfunction leading to nausea, vomiting, epigastric pain, jaundice, oliguria, and a hemorrhagic diathesis. Yellow fever differs from all other viral causes of hepatitis by the fact that serum aspartate aminotransferase (AST) levels exceed those of alanine aminotransferase (ALT). Aminotransferase levels are proportional to the severity of disease.16 Direct bilirubin levels range between 5 and 10 mg/dl, with levels being higher in those with fatal infection than in recovering patients. The diagnosis of yellow fever is confirmed by the serologic demonstration of specific IgM by enzyme-linked immunosorbent assay (ELISA), by polymerase chain reaction (PCR), or by isolation of the virus from blood. Liver biopsy is not recommended because of a high risk for hemorrhage.
The outcome of infection with yellow fever virus is determined during the second week after onset, when many patients recover rapidly, but between 20% and 50% of the patients who have progressed to the stage of intoxication will ultimately die of circulatory shock. Convalescence may be associated with fatigue for several weeks, and in some cases jaundice and elevated aminotransferase levels may persist for months.
Liver pathology varies according to the stage of the disease. In fatal cases, approximately 80% of hepatocytes undergo coagulative necrosis. The midzone of the liver lobule is affected, with sparing of cells neighboring the central vein and portal tracts.17 Very high viral loads have been detected in liver samples from patients who died, and viral antigen is located at the midzone of the hepatic lobule, thus indicating that this is the site of direct viral injury.18 Liver injury is characterized by eosinophilic degeneration and the presence of Councilman bodies. Rarely, intranuclear inclusion bodies (Torres bodies) are present. Fatty changes may be prominent. In specimens from surviving patients, the liver shows ballooned hepatocytes and regenerative hyperplasia with multiple multinucleated hepatocytes and some portal inflammation consisting mainly of CD4+ T cells and smaller numbers of natural killer and CD8+ T cells.19, 20 Biopsy specimens from survivors during the recovery phase may show a nonspecific acinar hepatitis.21 However, the hepatic reticular architecture is not disrupted, and in nonfatal cases healing is complete without any residual postnecrotic fibrosis.
Antiviral activity against yellow fever has been demonstrated for several nucleosides and plant alkaloids.22 Ribavirin inhibits yellow fever virus in vitro, but only at extremely high concentrations that cannot be achieved in vivo. Thus treatment is supportive. A highly active attenuated live vaccine is available that induces seroconversion rates of greater than 95% and provides a high level of protection. This vaccine should not be given to pregnant women and immunosuppressed individuals because of concern about the risk associated with live attenuated virus vaccines. Infrequently, two serious vaccine-related complications may also occur in immunocompetent persons: a form of encephalitis termed yellow fever–associated neurotropic disease and a syndrome resembling natural infection that is designated yellow fever vaccine–associated viscerotropic disease.
Lassa Fever
Lassa virus is an enveloped, single-stranded, bisegmented RNA virus that belongs to the Arenaviridae family. Its natural reservoir is the multimammated rat, Mastomys natalensis, which excretes the virus through urine, saliva, and other secretions.23 Humans presumably become infected directly via contact with infected rodent excreta through the fecal–oral route or inhalation of contaminated air.24 The risk of human–human transmission is low.25, 26 Serologic evidence demonstrates that the infection is widespread in western Africa, but clinically overt disease occurs in fewer than 10% and overall mortality is 1%. Increasing international travel has sporadically resulted in importation of Lassa fever to Western countries, which challenges the diagnostic skills of physicians.27
At onset, the infection is insidious, with fever, myalgia, headache, and malaise.28 More specific features include ulcerations in the oral cavity, bleeding from the gums, sore throat, cough, pleurisy, and watery diarrhea. There is lymphadenopathy and swelling of the liver and kidney, which become painful on palpation.29 Serum aminotransferase levels rise, but jaundice is not present. In Lassa fever the liver has a mottled appearance, and liver histology shows necrosis without inflammation. Single hepatocytes or groups of cells stain acidophilic and are found to harbor abundant arenaviruses under electron microscopy. There is no steatosis or cholestasis, whereas lipofuscin deposits are conspicuous.
ELISAs for Lassa virus antigen and immunoglobulins M and G (lgM and IgG)antibodies are sensitive and specific and in Africa have largely replaced indirect fluorescent antibody testing.30 In Western countries, the diagnosis can be established reliably by reverse transcription PCR (RT-PCR).31
Supportive therapy is often necessary. In addition, the antiviral drug ribavirin is effective against Lassa virus infection, but only if administered early in the course of illness.32 A study from Sierra Leone in patients at high risk for mortality confirmed the efficacy of ribavirin in the treatment of Lassa fever: only 1 of 20 patients died if intravenous ribavirin administered over a period of 10 days had been started within the first 6 days after the onset of fever, whereas mortality was 26% in the 43 patients whose treatment had begun later.33 However, because of its toxicity and teratogenicity, the need for intravenous application, and its expense, empiric therapy with ribavirin is not advised.33
Junin virus is another arenavirus that in Argentina can cause a viral hemorrhagic fever similar to Lassa fever.34 Ribavirin, as well as ribavirin-interferon combination therapy, has been attempted with some success in this disease.35
Ebola and Marburg Hemorrhagic Fever
Marburg and Ebola viruses are nonsegmented, negative-sense, single-stranded RNA viruses that belong to the Filoviridae virus family.36 They are among the most virulent pathogens and cause severe hemorrhagic fever and fulminant septic shock. No approved therapy is currently available to treat the devastating infections with Ebola and Marburg viruses.
All isolates of Marburg virus represent members of a single family, whereas Ebola isolates can be divided into different species (Zaire, Sudan, Ivory Coast, Reston, and probably Uganda 2007) that differ in their virulence.37, 38 The natural reservoirs of these viruses still remain a mystery, although it is suspected that both viruses are maintained in small animals, with bats heading the list of suspects.39, 40, 41, 42, 43 Retrospective analyses of African epidemics suggest that person–person transmission can occur through contact with virus-contaminated fluids (e.g., blood, vomitus, feces, urine).
Marburg virus fever was first detected in 1967, when people had come in contact with African green monkeys imported from Uganda. Mortality in this first outbreak was 25%. Meanwhile, natural outbreaks of Marburg virus and Ebola fever have been reported repeatedly from several western and central African regions (Fig. 34-1 ). Ebola and Marburg virus infections are still relevant. In 1989, monkeys imported into the United States from the Philippines died of Ebola virus infection. Outbreaks still occur in western Africa, and the risk of undiagnosed patients arriving in industrialized countries remains real. Information on current outbreaks is available on the World Health Organization website (www.who.int/topics/haemorrhagic_fevers_viral/en).
Fig. 34-1.
Localization of recent Ebola and Marburg virus outbreaks in Africa.
The disease caused by the various Marburg and Ebola virus isolates results in similar syndromes that differ only in severity and case fatality rates. The incubation period is 5 to 10 days but may exceed 20 days. Symptomatic persons have high levels of virus in their blood and must be considered contagious, so appropriate safety precautions should be taken.44 Marburg and Ebola infections typically begin with an abrupt onset of fever, chills, and general malaise. Other features include weakness, severe headache, pain in the muscles of the trunk and lower part of the back, nausea, vomiting, diarrhea, and abdominal pain.37, 45, 46 A nonproductive cough, pharyngitis, and a maculopapular rash on the upper half of the body are also frequent findings. Symptoms persist with worsening of prostration, stupor, and hypotension; disseminated intravascular coagulation eventually leads to conjunctival hemorrhages, easy bruising, and bleeding from venipuncture sites. Marked laboratory abnormalities include a striking leukopenia with immature granulocytes and abnormal lymphocytes, as well as thrombocytopenia. Filoviruses cause multifocal hepatic necrosis. Thus AST and ALT levels rise rapidly in the first days. Hypoproteinemia and proteinuria may also be present. The hepatic pathology is similar to that seen with Lassa virus infection: spotty to widespread necrosis of hepatocytes and minimal hepatic inflammation, but no cholestasis.47, 48, 49 Abundant filovirus particles are found in blood, phagocytes, endothelial cells, and hepatocytes. In lymph nodes, the reticular network is damaged.
Survivors of Marburg and Ebola hemorrhagic fever require prolonged convalescence and suffer from marked weakness, fatigue, and failure to regain weight. Marked sloughing of the skin and hair loss are commonly observed.50
Rift Valley Fever
Rift Valley fever is an acute vector-borne zoonotic disease caused by the Rift Valley fever virus, which belongs to the Bunyaviridae family and genus Phlebovirus. The virus was initially described in sheep and was first isolated from humans in Kenya.51 The disease is widespread in sub-Saharan Africa and also occurs in Egypt, Mauretania, Senegal, Saudi Arabia, and Yemen. Rift Valley virus is transmitted by several mosquito species or by direct contamination from infected animals. Patients with Rift Valley fever have symptoms and signs of an influenza-like illness. Fewer than 8% of patients progress to severe disease, including hepatitis, encephalitis, retinitis, and a generalized hemorrhagic syndrome.52
Crimean-Congo Hemorrhagic Fever
Crimean-Congo hemorrhagic fever (CCHF) is a tick-borne viral infection that occurs in parts of Africa, Asia, eastern Europe, and the Middle East.53 The virus belongs to the genus Nairovirus in the Bunyaviridae family. Isolates from different regions show considerable genetic diversity. Farmers and healthcare workers are particular at risk for this infection, but fortunately, CCHF develops in only one of five people infected. The disease has four stages: incubation, prehemorrhagic, hemorrhagic, and convalescence. Incubation takes 3 to 7 days, and then fever, headache, myalgia, and dizziness develop suddenly. Facial hyperemia, conjunctivitis, and occasionally diarrhea are also noted. This stage lasts 4 to 5 days. The hemorrhagic period is short (2 to 3 days). The most common bleeding sites are in the nose, the gastrointestinal tract, the genitourinary tract, and the respiratory tract. In survivors, the convalescence phase begins about 10 to 20 days after the onset of disease. RT-PCR is the method of choice for rapid laboratory confirmation of CCHF viral infection.54 Leukopenia and thrombocytopenia are consistent laboratory features, and raised levels of AST, ALT, and lactate dehydrogenase and prolonged coagulation tests are other common findings. Aminotransferase levels seem to correlate with the severity of disease.55, 56 Microscopic examination of liver tissue reveals variable degrees of hepatocellular necrosis with hemorrhage and Councilman bodies, fatty changes, Kupffer cell hyperplasia, and mild mononuclear portal inflammatory infiltrates.57
Treatment options are limited, but ribavirin has demonstrated antiviral activity against CCHF virus in vitro and in an animal model.58, 59 However, thus far, clinical effectiveness of ribavirin has been demonstrated only in observational studies.55, 60, 61, 62 In a recent outbreak, patients with severe disease were treated successfully with ribavirin for 10 days (30 mg/kg body weight as an initial loading dose, then 15 mg/kg every 6 hours for 4 days, and then 7.5 mg/kg every 8 hours for 6 days).53
Hantaviruses
Hantaviruses are prime examples of emerging viruses and are members of the genus Hantavirus within the Bunyaviridae family. They are negative-sense single-stranded RNA viruses that are carried primarily in rodents, which shed the virus in their urine, saliva, and feces. The virus is inhaled by humans as aerosols from dried rodent excreta or, in unusual circumstances, is transmitted via rodent bites.63 Hantaviruses exist in multiple serotypes worldwide, which differ in their virulence. Some are considered nonpathogenic, whereas certain isolates can produce two distinct severe syndromes in humans: Hantavirus cardiopulmonary syndrome, mostly caused by isolates in the Americas, and hemorrhagic fever with renal syndrome, caused by isolates (Seoul virus, Dobrava virus, Puumala virus, Hantaan virus) in Europe and Asia.64 In some instances, patients with Hantavirus hemorrhagic fever have suffered from severe acute hepatitis, whereas renal damage was rather mild.65, 66 Furthermore, a significantly increased prevalence of Hantavirus antibodies has been reported in patients with acute hepatitis of unknown cause from southwestern China67 and in patients with chronic hepatitis from Japan.68 However, the latter study failed to detect any Hantavirus antibodies in patients with acute hepatitis, thus implying a rather indirect pathogenetic role of Hantaviruses. Of note, Hantavirus infection has been observed to trigger acute exacerbations of autoimmune liver disease, and this mechanism has been proposed to contribute to community-acquired hepatitis.69 Nevertheless, the precise role of Hantavirus infections in human liver disease still awaits clarification. Analogous to other bunyaviruses, Hantavirus is sensitive to ribavirin, which, although not approved by the Food and Drug Administration, can be used as emergency treatment.70
Chikungunya Fever
Chikungunya is an arthropod-borne togavirus initially endemic to western Africa that in recent years has spread to the Indian Ocean islands and Southeast Asia.71 Although traditionally considered a disease of tropical and subtropical regions, in 2007 a local outbreak of chikungunya was even recorded in northern Italy.72 Because of international travel, chikungunya infections have also been exported from Southeast Asia and Indian Ocean islands to other Western countries. The infection begins abruptly with high fever, symmetric polyarthralgia, and macular or maculopapular rash.73 Pruritus and bullous skin lesions have also been described. Previously, chikungunya fever has been considered a self-limited disease. However, severe complications, including acute viral hepatitis and death, have been reported in recent outbreaks, particularly in elderly patients (>65 years) and people with chronic medical problems. Clinically, chikungunya fever must be differentiated from dengue fever, which shares many symptoms and features. Serology is the primary diagnostic tool in clinical practice, and chikungunya IgM antibodies become detectable by ELISA 5 days after the onset of symptoms. Viral culture and molecular techniques are valuable tools in a research setting. Although interferon alfa and ribavirin exhibit in vitro activity against chikungunya virus,74 an effective specific antiviral therapy does not exist. Thus treatment is primarily supportive. Chikungunya infection cannot currently be prevented by vaccination.
Viral Liver Disease Associated with Immunodeficiency
The establishment of organ transplantation as a routine therapeutic procedure, the growing use of anticancer chemotherapy, and the recent pandemics of human immunodeficiency virus (HIV) infection have resulted in increasing awareness of unusual manifestations of viral infections as complications of immunosuppression. Such infections occur either as acute disease during primary infection or as severe exacerbation of a latent viral persistence. Cytomegalovirus is the most common opportunistic pathogen that causes hepatitis in both patients with drug-induced immunosuppression and HIV-infected patients with advanced immunodeficiency, but the other members of the herpes family, as well as common community-acquired agents such as adenovirus, are also important causes in this setting.
Liver Disease Attributable to HIV
In approximately half of patients, a faint rash and an infectious mononucleosis–like syndrome develop 3 weeks to 6 months after a primary HIV infection. Several case reports have described a hepatitis-like syndrome in such patients that could not be explained by other concomitant infections.75, 76 Patients complained of vomiting, upper abdominal pain, and hepatomegaly. There was a rise in aminotransferases, but jaundice or elevated serum alkaline phosphatase was not present. The liver disease resolved spontaneously, and no histologic reports are available for this condition.
At autopsy, hepatomegaly was found in approximately two thirds of patients with acquired immunodeficiency syndrome (AIDS).77 In liver biopsy specimens, macrovesicular steatosis has been a common finding, which in rare instances could become excessive.78 HIV has repeatedly been detected in the liver, particularly in Kupffer cells and endothelial cells, both by in situ hybridization and by p24 immunohistochemical staining,79, 80, 81 thus suggesting that the liver might be an important site of HIV replication. A moderate chronic lymphocytic portal inflammation can be encountered in liver specimens and is thought to reflect HIV-associated reactive hepatitis. These nonspecific changes associated with HIV infection can then become superseded by further liver pathology caused by opportunistic infections and tumors (Fig. 34-2 ), illicit drug abuse, and currently, also the side effects of HIV antiretroviral therapy.82
Fig. 34-2.
Overview of opportunistic diseases associated with human immunodeficiency virus (HIV) infection and their relationship to progressing immunodeficiency.
AIDS, acquired immunodeficiency syndrome; CMV, cytomegalovirus; MAI, Mycobacterium avium-intracellulare complex.
The Herpesvirus Group
The herpesviruses form a large family of DNA viruses, and eight members cause disease in humans (Table 34-2 ). Herpes simplex virus (HSV), varicella-zoster virus (VZV), Epstein-Barr virus (EBV), cytomegalovirus (CMV), and human herpesvirus type 6 (HHV6) can directly affect the liver and are infections that in humans are usually acquired during childhood or adolescence. Herpesviruses persist lifelong and can reactivate and induce aggressive liver disease in immunosuppressed patients. Unlike the other herpesviruses, the prevalence of Kaposi sarcoma–associated virus, or HHV8, is more limited. HHV8 is found in saliva, which may serve as a source for transmission both sexually and probably also vertically from mother to child. Apart from Kaposi sarcoma, HHV8 can cause Castleman disease and body cavity lymphoma in severely immunosuppressed patients.
Table 34-2.
Human Herpesviruses
SYSTEMATIC NAME | COMMON NAME | HERPESVIRUS SUBFAMILY | ESTIMATED PREVALENCE | ROUTE OF TRANSMISSION |
---|---|---|---|---|
Human herpesvirus 1 | Herpes simplex virus 1 (HSV1) | α-Herpesvirinae | 75% to >95% | Oral secretions |
Close contact | ||||
Human herpesvirus 2 | HSV2 | α-Herpesvirinae | 4% to 95% | Genital secretions |
Close contact | ||||
Human herpesvirus 3 | Varicella-zoster virus (VZV) | α-Herpesvirinae | >90% | Aerosol |
Close contact | ||||
Human herpesvirus 4 | Epstein-Barr virus (EBV) | γ-Herpesvirinae | 70% to 95% | Oral secretions |
Human herpesvirus 5 | Human cytomegalovirus (CMV) | β-Herpesvirinae | 40% to 95% | Oral secretions |
Genital secretions | ||||
Human herpesvirus 6 | Human herpesvirus 6 (HHV6) | β-Herpesvirinae | >85% | Oral secretions |
Human herpesvirus 7 | Human herpesvirus 7 (HHV7) | β-Herpesvirinae | >85% | Oral secretions |
Human herpesvirus 8 | Kaposi sarcoma–associated herpesvirus | γ-Herpesvirinae | 10% to 25% | Oral secretions |
Genital secretions |
Herpes Simplex Virus
Primary herpes simplex infection produces characteristic oral (HSV1) or genital (HSV2) vesicular lesions on an erythematous base. The symptoms can be severe, with fever and malaise, but many primary infections are asymptomatic. Once primary infection has been established, the virus adopts a latent state and persists in the nerve cell bodies of the dorsal ganglia, from where it can reactivate under immunosuppression.
Fulminant hepatitis is a complication of both HSV1 and HSV2 infection.83 Organ transplantation and treatment of hematologic malignancies are the most frequent underlying predispositions.84, 85, 86 Other individuals at risk include neonates, patients taking steroids, HIV-infected patients, and patients with cancer or myelodysplastic syndromes.83, 85, 86, 87, 88, 89, 90 Fatal HSV-related hepatitis has also rarely been reported in immunocompetent adults.91 HSV-related hepatitis has high mortality (>80%) and resembles septic endotoxic shock; jaundice is not always present.85, 87 Clinical features include fever, anorexia with nausea and vomiting, abdominal pain, leukopenia, and coagulopathy. Typical oral or genital vesicular lesions occur in only about a third of patients.83 Some patients also have disseminated extrahepatic involvement of the lung, lymph nodes, spleen, and adrenal glands. The diagnosis of HSV-related hepatitis must be established rapidly via detection of HSV by either viral isolation,92 immunofluorescence staining, or preferentially, PCR92, 93; serologic assays have rather limited application.
In fatal cases the liver is enlarged and congested at autopsy and has a mottled appearance with multiple white and yellow foci. Microscopy reveals irregular parenchymal necrosis with little inflammation. At the margins of necrotic areas multinucleated hepatocytes may be found, and hepatocytes may contain purple nuclear inclusion bodies with a surrounding halo.88
After organ transplantation, none of the patients survived once the liver was diffusely involved, whereas three of seven patients with more focal disease had survived with acyclovir therapy.85 Prompt systemic treatment with acyclovir has also been shown to reduce HSV-associated morbidity and the risk for serious complications in HIV-infected patients.94 In addition, anti-HSV prophylaxis with acyclovir at organ transplantation has markedly reduced HSV reactivation after surgery.95, 96 Acyclovir resistance occurs in about 5% of immunocompromised patients and is negligible (<0.5%) in immunocompetent subjects.97 Valacyclovir, a prodrug of acyclovir, and famciclovir, a prodrug of penciclovir, have similar antiviral mechanisms as acyclovir, and thus HSV isolates resistant to acyclovir are also resistant to these drugs.98 Cidofovir and foscarnet are alternative choices to treat acyclovir-resistant HSV, but side effects are greater with these drugs than with acyclovir.99
Varicella-Zoster Virus
Varicella-zoster virus causes chickenpox, as well as shingles when latent infection is reactivated. In a first viremic phase after infection, VZV replicates in the epithelia of the gut, respiratory tract, liver, and endocrine glands. A secondary viremia leads to infection of the skin, which results in the usual rash. Liver disease is rare and limited to patients with severe immunodeficiency. Anticancer chemotherapy and corticosteroid treatment are risk factors for this complication.100 In severe VZV infection, hepatic lesions are similar to herpes simplex hepatitis, with massive liver necrosis and inclusion bodies but little inflammation. Electron microscopy and immunohistochemistry reveal abundant VZV. The diagnosis is best made by PCR,93 and treatment and prophylaxis are similar to that for HSV, with acyclovir also being effective against VZV. In addition, VZV has been reported to trigger severe autoimmune hepatitis.101
Epstein-Barr Virus
Epstein-Barr virus is the agent that accounts for 90% of acute infectious mononucleosis syndromes. It persists lifelong in a latent state, which results from a dynamic interplay between viral evasion strategies and the host's immune responses. However, unlike other herpesviruses, reactivation-associated disease is not a prominent feature in chronic EBV disease. Still, EBV is a potent cause in a variety of malignancies, such B- and T-cell lymphomas, Hodgkin lymphoma, and nasopharyngeal carcinoma. In transplant patients, EBV has been associated with an aggressive lymphoproliferative disease.
EBV is shed in oral secretions, and most primary EBV infections originate in the oropharynx. Based on genetic differences in the EBV nuclear antigen-3 genes, two types of EBV have been identified102 that show divergent prevalence throughout the world: in Western countries, EBV1 is 10 times more prevalent than EBV2, whereas in Africa, the two EBV genomes are distributed equally.103
Because few pregnant women are susceptible, intrauterine EBV infection is rare but in isolated cases may lead to diverse congenital anomalies, including biliary atresia.104 In infants and young children, primary infection is common and frequently asymptomatic, whereas in adults, it results in the infectious mononucleosis syndrome, which begins with malaise, headache, and low-grade fever before more specific symptoms such as pharyngitis/tonsillitis, swelling of the cervical lymph nodes, and moderate to high-grade fever develop.105 Patients have peripheral blood lymphocytosis with characteristic large abnormal lymphocytes in their blood smears. Nausea, vomiting, and anorexia are common findings and probably reflect the mild hepatitis that accompanies infectious mononucleosis fever in approximately 90% of patients. Liver histology shows diffuse lymphocytic infiltrates in the sinusoids, and focal apoptotic hepatocytes can occasionally be seen (Fig. 34-3, A ).106, 107 These infiltrates may be atypical and must then be carefully differentiated from leukemia/lymphoma. Nonnecrotic hepatic granulomas are occasionally detected in patients with infectious mononucleosis. Splenomegaly is found in about half of patients, but hepatomegaly and jaundice are uncommon findings. The vast majority of patients recover over a period of 2 to 4 weeks, but fatigue may persist for several months after infection.
Fig. 34-3.
Histologic changes in the liver associated with Epstein-Barr virus (A) and cytomegalovirus (B and C) infections.
A, Prominent sinusoidal infiltration of lymphocytes into the sinusoidal space. B, Cytomegalovirus inclusion bodies (arrow) and immunohistochemical nuclear staining for cytomegalovirus antigen. C, Microabscesses in early cytomegalovirus infection.
Patients with X-linked lymphoproliferative disease, which is caused by a mutation in the SH2D1A gene on the X chromosome, are particularly vulnerable to EBV108 and may suffer fatal infections with extensive liver necrosis.109 In patients with severe immunodeficiency, lymphomatoid granulomatosis is a further unusual complication of EBV infection that leads to granuloma formation in multiple organs, including the liver, and may require interferon alfa antiviral therapy.110 An EBV-associated lymphoproliferative disorder with hepatic infiltration of immunoblasts111 and a hemophagocytic syndrome112 have also been reported in patients with HIV infection.
EBV is also the major causative agent of the so-called posttransplant lymphoproliferative disease (PTLD), which after organ transplantation may result in lymphocytic infiltration of the liver and other organs ranging from benign polyclonal B-cell proliferation to malignant B-cell lymphoma.113 PTLD occurs more commonly in children than in adults and is related to the type and degree of immunosuppression. It is a particularly common complication in EBV-negative individuals who receive a graft from an EBV-positive donor, with primary EBV infection developing as a result of immunosuppression.114 Clinical suspicion of EBV infection is confirmed by detection of heterophilic or EBV-specific antibodies in patients with infectious mononucleosis and quantitative PCR assays in those with lymphoproliferative disorders.115, 116
Treatment of EBV infection is primarily supportive. Corticosteroid therapy can ameliorate the symptoms but is not generally recommended because EBV infection is usually a self-limited disease and there are theoretic concerns of suppression of immune responses with a viral infection, which can potentially cause malignant disease. Nevertheless, corticosteroids should be considered in individuals with life-threatening complications such as imminent liver failure. Acyclovir inhibits EBV DNA polymerase, and antiviral therapy with this drug has shortened viral shedding but failed to demonstrate a convincing clinical benefit even in patients with severe acute EBV infection.117 Acyclovir does not affect latent EBV infection. Thus anticancer chemotherapy,118, 119 B-cell monoclonal antibodies,120, 121, 122 and if possible, a reduction in immunosuppression123 are needed to treat EBV-related lymphoproliferative disorders, but antiviral therapy in general is not effective.
Cytomegalovirus
In immunocompetent hosts, CMV infection is asymptomatic or causes only transient, minimally symptomatic acute disease. Newborns and immunocompromised patients, such as those with HIV infection, cancer, and solid organ or bone marrow transplantation, are commonly infected with CMV and may be susceptible to the development of serious disease.
In about 10% of immunocompetent subjects, primary CMV infection produces an infectious mononucleosis–like syndrome that is associated with elevated serum aminotransferase levels and a mild hepatitis. Liver histology may show focal hepatocyte and bile duct damage with lymphocytic infiltration into the sinusoids.124 In other patients, epithelioid granulomas without necrosis were present, but CMV inclusion bodies or CMV immunostaining were rarely detectable.124, 125
Congenital CMV infection is observed in fewer than 2% of newborns and commonly occurs when mothers have primary CMV infection or reactivation of CMV during pregnancy. Approximately 10% of such neonates have symptoms at birth, including hepatosplenomegaly and jaundice.126 Fetal CMV infection has also been associated with obstructive biliary disease and neonatal hepatitis with giant cell transformation, cholestasis, and viral inclusion bodies.127 In CMV infection of the liver, endothelial cells, Kupffer cells, and hepatocytes become swollen and may contain basophilic granules in their cytoplasm. A typical intranuclear amphophilic inclusion body surrounded by a clear halo resembling an owl's eye may also be present (see Fig. 34-3, B). Both nuclear and cytoplasmic inclusions are full of virions.128
CMV-related liver disease is the most common cause of acute viral hepatitis in patients after organ transplantation. Infection may result from reactivation of endogenous virus because of immunosuppression, transmission from the transplanted organ of a CMV-positive donor, or transfusion of blood and blood products. In liver transplantation, most CMV disease occurs after 1 and 4 months, and CMV infection is also discussed as a potential risk factor for the subsequent development of acute and chronic rejection. On the other hand, rejection therapy with corticosteroid boluses may trigger reactivation of endogenous CMV. Liver biopsy specimens from patients with posttransplant CMV hepatitis usually show scanty CMV inclusion bodies, which may be associated with small foci of necrosis and inflammation (microabscesses) (see Fig. 34-3, C).
Although reactivation of CMV is also a common severe complication in HIV-positive patients with advanced immunodeficiency (usually CD4+ counts <50/µl), hepatic involvement seems to be rather minor.129 Occasionally, CMV causes severe bile duct necrosis, and it is also a major cause of HIV cholangiopathy, a sclerosing cholangitis that is encountered rarely in patients with terminal HIV-related immunodeficiency.130, 131, 132
De novo appearance of CMV antibodies of the IgM class or a four-fold rise in IgG antibodies is considered indicative of active CMV infection in immunocompetent individuals. However, serology is unreliable in immunocompromised patients and is being replaced by quantitative molecular DNA amplification assays.133, 134, 135, 136 Currently, most transplant centers perform CMV surveillance by weekly quantitative determination of CMV DNA and also administer hyperimmune antibodies or antiviral drugs for CMV prophylaxis to transplant recipients at high risk for acquiring CMV disease.137, 138 However, CMV infection and disease may still develop. In patients with immunodeficiency, CMV hepatitis should be treated promptly. At present, intravenous ganciclovir or oral valganciclovir for 3 weeks is the treatment of choice. Treatment can be continued in reduced doses as chemoprophylaxis if prolonged immunosuppression is anticipated.139 Fortunately, ganciclovir resistance seems to be a rather rare event,140 so the toxic alternatives cidofovir and foscarnet can largely be avoided.
Human Herpesvirus 6 and Human Herpesvirus 7
There are two variants of HHV6, HHV6A and HHV6B, which infect not only T cells but also other cells types expressing the CD46 receptor.141 Though genetically clearly distinct from HHV6, HHV7 is another β-herpesvirus that shares many features with HHV6. Primary infection with either virus commonly occurs at a young age and can lead to a febrile illness known as exanthema subitum or roseola infantum.142 Pityriasis rosea reflects primary infection with HHV7. Although the spectrum of diseases caused by HHV6 and HHV7 later on is not fully known, these viruses have been linked to a variety of different syndromes such as encephalitis, multiple sclerosis, pneumonitis, an infectious mononucleosis–like condition, and postinfectious drug hypersensitivity, as well as lymphoproliferative disorders and systemic disease in immunocompromised patients.143
HHV6 can also cause hepatitis, which has occasionally resulted in liver failure in neonates and young infants.144, 145 Moreover, HHV6 DNA and antigens have been detected in a significant proportion of children and adults who underwent transplantation for acute liver failure of unknown cause.146, 147, 148, 149 Of note, in two patients with HHV6 infection and imminent liver failure, HHV6 infection was controlled successfully with valganciclovir and with liver transplantation plus ganciclovir, respectively.150 Hepatitis caused by HHV6 and HHV7 also complicates organ transplantation.151, 152, 153 HHV6-induced acute liver failure and co-infection with hepatitis B and C viruses have been identified as risk factors for reactivation of HHV6 in patients after liver transplantation.154, 155 Of note, HHV6 reactivation seems to significantly increase mortality in living related liver transplant recipients.155 HHV6 and HHV7 can reactivate each other156, 157 and can also reactivate CMV infection and lead to symptomatic disease in liver transplant patients.158 In addition, HHV6 has been associated with autoimmunity and postinfantile giant cell hepatitis,159, 160 as well as giant cell transformation of bile duct cells.161
Seroconversion with at least a four-fold rise in IgG antibody titer between paired samples is considered diagnostic of HHV6 infection, but serology does not distinguish between HHV6A and HHV6B variants and may also be cross-reactive with HHV7. HHV6 IgM antibodies develop within a week after infection but are an unreliable marker because approximately 5% of healthy subjects may have a positive IgM test at any given time.162 HHV6 can be detected in tissue samples by virus-specific monoclonal antibodies, but the preferred method for diagnosing HHV6 and HHV7 infection is by quantitative DNA amplification assays.163, 164, 165
In most immunocompetent patients, HHV6 and HHV7 cause a benign self-limited infection that does not require specific antiviral treatment. Nevertheless, foscarnet is active against HHV6A, HHV6B, and HHV7 in vitro, whereas unlike HHV6B, both HHV6A and HHV7 may be relatively resistant to ganciclovir.166, 167, 168, 169 Cidofovir may be a therapeutic alternative, but some resistant HHV6 isolates have been identified.170 Although in vitro studies and anecdotal reports in the transplant setting seem to suggest some beneficial effects in human infections,150, 169 antiviral therapy against HHV6 or HHV7 has not yet been evaluated in controlled trials.
Human Herpesvirus 8
HHV8 is a γ-herpesvirus that has the potential for malignant transformation. Although primary HHV8 infection can cause rash and fever in children and immunocompromised individuals, the onset of HHV8-related diseases usually occurs several years after the acquisition of HHV8. Kaposi sarcoma, body cavity lymphoma, and multicentric Castleman disease are the typical manifestation of HHV8 infection, but bone marrow aplasia and multiple myeloma have also been described in association with this infection.171 T lymphocytes play an important role in the control of HHV8 infection, which explains why the incidence of HHV8-related diseases is increased in patients with a transplant or AIDS. This concept is also supported by the observation that regression of Kaposi sarcoma is achieved when immunosuppression is reduced in patients with a transplant172, 173 and when highly active antiretroviral therapy (HAART) has improved immune function in subjects with AIDS.174 Other risk factors for Kaposi sarcoma, the predominant manifestation of HHV8-associated disease, are multiple homosexual contacts in HIV-infected men175 and, in the transplant setting, male sex, old age, and lung transplantation.176
In autopsy studies, Kaposi sarcoma has involved the liver in approximately 20% of subjects with AIDS, but its prevalence is declining with HAART. Hepatic Kaposi sarcoma is usually part of widespread cutaneous and visceral disease but may rarely be the primary manifestation.177 Fulminant hepatic Kaposi sarcoma has also been observed in the first few weeks after liver transplantation (Fig. 34-4, A to E ). Macroscopically, dark red tumors 0.5 to 2 cm in diameter may be seen on the skin, the liver capsule, and the parenchyma. At the microscopic level, the typical lesion is a mesh of spindle cell–like tumour cells and dilated thin-walled vessels.178 Diffuse plasma cells, infrequent mitosis, and clusters of intracytoplasmic eosinophilic inclusions resembling small erythrocytes are further characteristic features. Hepatosplenomegaly, fever, and weight loss are typical features of multicentric Castleman disease, a noncancerous proliferation of B lymphocytes (see Fig. 34-4, F). Lymphocytes in patients with multicentric Castleman disease and Kaposi sarcoma seem to cooperate with each other, and thus the two HHV8-related lesions can occasionally be found within the same lymph node.179, 180
Fig. 34-4.
Human herpesvirus 8 (HHV8)-associated manifestations.
A, Cutaneous Kaposi sarcoma developing in a surgical scar 4 weeks after liver transplantation. B, Detection of HHV8 latency-associated nuclear antigen-1 in a skin section from the patient in A. C, Endoscopic retrograde cholangiopancreatography demonstrates compressed and partially blocked biliary ducts as a result of multifocal hepatic tumor infiltration (same patient as in A). D, Liver histology illustrating typical Kaposi sarcoma with abundant spindle cells and dilated vascular spaces (same patient as in A). E, High-power field of hepatic Kaposi sarcoma. F, Multicentric Castleman disease in a perihepatic lymph node. Histology shows many atypical follicle-like structures and abnormal vessels. G, Plasma cell–like lymphoma cells in an HIV-infected patient with HHV8-associated body cavity lymphoma.
Ascites and pleural effusions may initially raise suspicion of some underlying liver disease in patients with HHV8-related body cavity lymphoma, but they can be differentiated by the presence of abundant lymphoma cells in aspirated fluid (see Fig. 34-4, G). HHV8 can also cause solid organ lymphoma, which in the liver leads to diffuse sinusoidal accumulation of HHV8-infected B lymphocytes.181
Reconstitution of immune function is a primary goal in the treatment of HHV8-associated diseases and, apart from HAART in HIV infection, may be achieved with the use of antiproliferative m-TOR (mammalian target of rapamycin) inhibitors for immune suppression in transplantation182, 183 or immune stimulation with imiquimod and interferon alfa.184, 185 Chemotherapy with liposomal anthracyclines or paclitaxel for Kaposi sarcoma186, 187 and rituximab for Castleman disease and lymphoma are further potent treatment options.188 Ganciclovir, cidofovir, foscarnet, adefovir, and lobucavir, but not acyclovir, show some in vitro activity in blocking replication of HHV8-infected cell lines, and in a double-blind, placebo-controlled crossover trial, valganciclovir reduced oropharyngeal shedding of HHV8 by 80%.189
Adenoviruses
Adenoviruses have a worldwide distribution and cause febrile diseases in infants and young children. More than 50 serotypes can be distinguished and are further subdivided into six subgroups, A through F. Typical syndromes include conjunctivitis, upper respiratory tract infections such as pharyngitis and coryza, pneumonia, and otitis media. In young children, an acute diarrheal illness is caused by subgroup F type 40 and 41 adenoviruses. Adenoviruses can persist in human tissue over prolonged periods190, 191 and can cause a variety of clinical syndromes in immunocompromised individuals, including serious hepatitis.192 Adenoviral hepatitis occurs in congenital and acquired immunodeficiency syndromes and may be fatal in these individuals.193, 194, 195, 196 Adenoviral hepatitis is a particular problem in pediatric liver transplantation and was identified in 14 (3%) of 484 pediatric liver transplant recipients197; 6 of the 14 patients died, 4 required retransplantation, and only 4 patients recovered with a decrease in immunosuppressive therapy. Transmission of latent adenovirus with the donated organ seems to be a potential risk factor for this complication.198 The signs and symptoms of adenoviral hepatitis resemble those of HSV infection and consist of massively elevated aminotransferase levels and severe coagulopathy. There are extensive areas of liver cell necrosis with little inflammation and intranuclear inclusion bodies (Fig. 34-5, A to D ). To date, a proven therapy for adenoviral hepatitis does not exist, but ribavirin may be helpful in selected cases.199
Fig. 34-5.
Fulminant adenoviral hepatitis in an HIV-infected woman.
A, Computed tomographic scan illustrating extensive focal necroses in the liver (arrow). B, Necrosis and enlarged hepatocytes with adenoviral inclusions (same patient as in A). C, Immunohistochemistry demonstrates the presence of abundant adenoviral antigens (same patient as in A). D, Intracellular adenoviral virions seen on electron microscopy (same patient as in A).
Liver Disease Associated with Systemic Viral Infections
The liver can be affected as part of a generalized infection with viruses that primarily target tissues other than liver, such as adenovirus and influenza. Liver involvement can range from asymptomatic deranged biochemistry to fulminant hepatic failure. Loss of immune control is considered responsible for hepatitis associated with opportunistic viral infections, and rather similar mechanisms may operate in severe acute respiratory syndrome (SARS)-associated hepatitis, which is characterized by focal lobular lymphocytic infiltrates. Finally, “collateral damage” has been proposed in influenza infection and reflects expansion of virus-specific CD8+ T cells generated outside the liver that may trigger T-cell–dependent hepatitis even when viral antigens are apparently absent in the liver.200 Kupffer cells play a pivotal role in this process because they can take up and present viral antigens, coordinate lymphocyte recruitment, or possibly become activated by lymphocytes to induce hepatocellular apoptosis.201 Because T-cell expansion occurs with many viral infections,202 this bystander mechanism may contribute to hepatitis in many extrahepatic infections, with important implications for liver pathobiology.
Influenza
Influenza viruses represent three genera in the Orthomyxoviridae family. Generally, influenza A viruses are associated with more severe disease in humans than are influenza B and C viruses. Influenza A is further subdivided with respect to genetic variation in its hemagglutinin (H) and neuraminidase (N) genes. Influenza viruses commonly cause a self-limited acute respiratory infection manifested as fever, rhinorrhea, sore throat, and occasionally, gastrointestinal symptoms. Therefore aminotransferase levels are not monitored routinely. In the 2004 H1N5 influenza outbreak, however, about 60% of patients with pneumonia had deranged liver function test values and gastrointestinal symptoms such as vomiting, abdominal pain, or diarrhea on initial evaluation.203 Of note, among other extrahepatic abnormalities, autopsy revealed hepatic centrolobular necrosis, although neither viral antigen detection nor RT-PCR could provide evidence of viral liver disease.204 Thus liver damage in patients dying of influenza has been considered immune mediated, in particular because high cytokine levels were detected.205 Of note, in 4 of 15 volunteers infected intranasally with influenza A/Kawasaki/86 (H1N1), markedly elevated aminotransferase levels also transiently developed.200 This experimental observation is remarkable because the rise in liver enzymes occurred after pyrexia had settled, thus suggesting that the liver damage was mediated by the host's immune response rather than viral infection of the liver. It has been proposed that immune-mediated liver damage may also be the cause of elevated aminotransferases in other viral respiratory infections such as respiratory syncytial virus,206 although cardiovascular effects and hepatic ischemia must be considered in these severely ill patients as well.207
Coronavirus (Severe Acute Respiratory Syndrome)
A novel coronavirus (SARS-coronavirus [SARS-CoV]), causes SARs, and it resulted in an outbreak of severe infection of the lung and gastrointestinal tract in the Far East and Canada.208, 209 There was also involvement of other organs; approximately 25% of patients had elevated liver enzymes at the onset of infection, and elevated levels developed later in an additional 45% of patients with normal liver enzymes at initial evaluation, so overall, up to 70% of patients had elevated liver enzyme levels during their illness.210, 211, 212, 213, 214, 215 Jaundice was observed in less than 10% of patients. In most patients, aminotransferase levels started to rise toward the end of the first week and peaked at the end of the second week. With resolution of SARS, aminotransferases normalized spontaneously in the majority of patients. Severe liver damage (ALT more than five times the upper limit of normal) was observed more frequently in male patients and those with significant other co-morbid conditions or elevated serum creatinine levels.215 The role of concomitant hepatitis B in disease severity is controversial.214, 215 However, there was a close relationship between the severity of hepatic dysfunction and degree of pulmonary damage and the outcome of SARS.214 Thus high ALT levels appeared to be an independent predictor of more severe disease and a worse outcome.
Histopathologic studies revealed marked apoptosis of hepatocytes and conspicuous (presumably compensatory) mitotic activity.216 Hepatocytes also showed some ballooning, and mild to moderate lymphocytic infiltrates were described in the portal tracts and liver lobules. Although SARS-CoV RT-PCR was consistently positive in liver specimens, immunohistochemistry and electron microscopy failed to detect viral antigens and viral particles in several biopsy and postmortem studies.216, 217, 218 Thus only small amounts of SARS-CoV seem to be present in the liver, and analogous to influenza infection, the host's immune response may contribute to the liver damage.219 In the recent SARS outbreak, both ribavirin and Kaletra (baby-dose ritonavir/lopinavir) were used as experimental therapy with limited success.
Measles (Rubeola)
Measles is caused by Morbillivirus of the Paramyxoviridae family. It is an acute febrile illness with a typical rash that is currently preventable by vaccination. Measles is associated with diverse complications such as pneumonitis or subacute sclerosing panencephalitis and causes significant mortality in third-world countries and in patients with immunodeficiency.220 Liver dysfunction has increasingly been recognized as a complication of measles in children and adults221, 222, 223 and may become a prominent feature in patients with atypical measles and partial immunity to the measles virus.224, 225 Hepatic abnormalities have been noted to occur more frequently in adult patients with primary infection (up to 66%) than in children.226 Two patterns of hepatic dysfunction are encountered: asymptomatic elevations of aminotransferases, which resolve within a few days, and rarely, prolonged cholestasis and jaundice, which may appear when measles begins to recede.227 In liver tissue, steatosis, portal inflammation, and focal necrosis have been reported.228 Viral inclusions and giant cells are rarely seen but have been observed in a child with congenital immunodeficiency.229 In addition, measles has been incriminated in triggering autoimmune hepatitis.230 Paramyxovirus particles have also been reported in several patients with syncytial giant cell hepatitis.231, 232, 233, 234 However, the very nature of these particles has thus far remained elusive.
Rubella
Childhood rubella is apparently not associated with significant liver disease. Nonetheless, this virus has been proposed as a cause of neonatal giant cell hepatitis, which may also be accompanied by necrosis, cholestasis, and lymphocytic infiltrates on histologic examination.235
Enteroviruses
Group B coxsackievirus infection can cause liver disease as part of a serious multisystem infection in young children and also rarely in adults.236, 237, 238 Liver histology shows hemorrhagic necrosis in neonates, whereas in adults, swollen hepatocytes, bile stasis, and a mixed infiltrate of mononuclear and polymorphic leukocytes in the portal tracts and sinusoids have been observed.
Parvovirus
Human parvovirus B19 is a nonenveloped, single-stranded DNA virus that belongs to the Erythrovirus genus of the Parvoviridae family. Humans are the only known host, in whom it can produce a wide range of different syndromes. Most immunocompetent people with B19 infection will be asymptomatic or suffer only nonspecific malaise, muscle pain, and fever, which in the second week may be followed by rash, arthralgia, pancytopenia, and edema in about a quarter of infected patients.239, 240 Erythema infectiosum occurs in school-aged children and occasionally also in adults. Arthropathy and aplastic anemia are further complications in adults. Fetal infection during gestation is a cause of hydrops fetalis, which is associated with marked damage to the liver241; hepatocytes have ballooned, swollen nuclei that harbor eosinophilic nuclear inclusion bodies. In addition, erythroid-myeloid precursors are greatly expanded in the sinusoids. Parvovirus B19 has also been identified as a cause of acute hepatitis242 and has been incriminated as the underlying cause in acute liver failure with anemia.243 However, a subsequent study could not confirm parvovirus B19 as a cause of fulminant hepatic failure.244
Torquetenovirus and other Anelloviruses
Torquetenovirus (TTV) was initially identified as a novel virus in three of five patients in whom posttransfusion hepatitis developed but were found to be negative for all known hepatitis viruses.245 TTV is classified in the genus Anellovirus, which is not attached to any family. Torquetenominivirus and related viruses with smaller genomes, provisionally designated as small anelloviruses, are further members of this genus. TTV can be subdivided into five phylogenetic groups, 1 to 5, and co-infection with several genotypes is common. Some TTV variants have their own designation, such as SEN virus, named after the patient from whom it had been isolated.
Soon after its discovery, it became clear that TTV viremia occurs frequently in the healthy general population worldwide. Thus the clinical significance of TTV infections remains uncertain. Inoculation of chimpanzees with TTV led to viremia but did not cause hepatitis.246 Moreover, several studies on TTV transmission failed to establish any consistent relationship between TTV viremia and elevated serum aminotransferase levels.247, 248, 249 Nevertheless, it cannot be excluded that TTV is responsible for acute hepatitis in some subsets of patients,247, 250 and if so, TTV seems to cause a rather mild form of hepatitis.251
Likewise, most studies also could not confirm any consistent associations between persistence of TTV and the presence of biochemical and histologic hepatic abnormalities in a variety of different clinical settings.248, 252, 253, 254 TTV infection does not seem to trigger autoimmune hepatitis,255 and a case-control study failed to establish TTV as an independent risk for factor hepatocellular carcinoma.256
Taken together, it seems most likely that the initial reports of an increased TTV prevalence in patients with chronic liver disease257, 258 reflect shared risk factors for infection and liver damage rather than TTV as the cause of the liver disease.
Abbreviations
- AIDS
acquired immunodeficiency syndrome
- ALT
alanine aminotransferase
- AST
aspartate aminotransferase
- CCHF
Crimean-Congo hemorrhagic fever
- CMV
cytomegalovirus
- DEN1 to DEN4
dengue virus serotypes 1 through 4
- DHF
dengue hemorrhagic fever
- DSS
dengue shock syndrome
- EBV
Epstein-Barr virus
- ELISA
enzyme-linked immunosorbent assay
- HAART
highly active antiretroviral therapy
- HHV
human herpesvirus
- HIV
human immunodeficiency virus
- HSV
herpes simplex virus
- IgG
immunoglobulin G
- IgM
immunoglobulin M
- PTLD
posttransplant lymphoproliferative disease
- RT-PCR
reverse transcription polymerase chain reaction
- SARS
severe acute respiratory syndrome
- TTV
torquetenovirus
- VZV
varicella-zoster virus
Key References
- Alexopoulou A. A fatal case of postinfantile giant cell hepatitis in a patient with chronic lymphocytic leukaemia. Eur J Gastroenterol Hepatol. 2003;15:551–555. doi: 10.1097/01.meg.0000050026.34359.7c. (Ref.234) [DOI] [PubMed] [Google Scholar]
- Al-Hoamoudi WK. Severe autoimmune hepatitis triggered by varicella zoster infection. World J Gastroenterol. 2009;15:1004–1006. doi: 10.3748/wjg.15.1004. (Ref.101) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alsop Z. Ebola outbreak in Uganda “atypical,” say experts. Lancet. 2007;370:2085. doi: 10.1016/s0140-6736(07)61880-7. (Ref.38) [DOI] [PubMed] [Google Scholar]
- Babel N. Development of Kaposi's sarcoma under sirolimus-based immunosuppression and successful treatment with imiquimod. Transpl Infect Dis. 2008;10:59–62. doi: 10.1111/j.1399-3062.2007.00239.x. (Ref.184) [DOI] [PubMed] [Google Scholar]
- Bae HG. Analysis of two imported cases of yellow fever infection from Ivory Coast and The Gambia to Germany and Belgium. J Clin Virol. 2005;33:274–280. doi: 10.1016/j.jcv.2004.12.001. (Ref.18) [DOI] [PubMed] [Google Scholar]
- Barnett ED. Yellow fever: epidemiology and prevention. Clin Infect Dis. 2007;44:850–856. doi: 10.1086/511869. (Ref.15) [DOI] [PubMed] [Google Scholar]
- Barozzi P. Indirect antitumor effects of mammalian target of rapamycin inhibitors against Kaposi sarcoma in transplant patients. Transplantation. 2009;88:597–598. doi: 10.1097/TP.0b013e3181b15d56. (Ref.183) [DOI] [PubMed] [Google Scholar]
- Bonnafous P. Characterization of a cidofovir-resistant HHV-6 mutant obtained by in vitro selection. Antiviral Res. 2008;77:237–240. doi: 10.1016/j.antiviral.2007.12.004. (Ref.170) [DOI] [PubMed] [Google Scholar]
- Briolant S. In vitro inhibition of chikungunya and Semliki Forest viruses replication by antiviral compounds: synergistic effect of interferon-alpha and ribavirin combination. Antiviral Res. 2004;61:111–117. doi: 10.1016/j.antiviral.2003.09.005. (Ref.74) [DOI] [PubMed] [Google Scholar]
- Booth CM. Clinical features and short term outcome of 144 patients with SARS in the greater Toronto area. JAMA. 2003;289:2801–2809. doi: 10.1001/jama.289.21.JOC30885. (Ref.211) [DOI] [PubMed] [Google Scholar]
- Bower M. Brief communication: rituximab in HIV-associated multicentric Castleman disease. Ann Intern Med. 2007;147:836–839. doi: 10.7326/0003-4819-147-12-200712180-00003. (Ref.188) [DOI] [PubMed] [Google Scholar]
- Cacheux W. HHV-6–related acute liver failure in two immunocompetent adults: favourable outcome after liver transplantation and/or ganciclovir therapy. J Intern Med. 2005;258:573–578. doi: 10.1111/j.1365-2796.2005.01567.x. (Ref.150) [DOI] [PubMed] [Google Scholar]
- Caliendo AM. Comparison of molecular tests for detection and quantification of cell-associated cytomegalovirus DND. J Clin Microbiol. 2003;41:3509–3513. doi: 10.1128/JCM.41.8.3509-3513.2003. (Ref.135) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Campistol JM, Schena FP. Kaposi's sarcoma in renal transplant recipients—the impact of proliferation signal inhibitors. Nephrol Dial Transplant. 2007;22(Suppl 1):17–22. doi: 10.1093/ndt/gfm089. (Ref.182) [DOI] [PubMed] [Google Scholar]
- Capello D. Analysis of immunoglobulin heavy and light chain variable genes in post-transplant lymphoproliferative disorders. Hematol Oncol. 2006;24:212–219. doi: 10.1002/hon.791. (Ref.114) [DOI] [PubMed] [Google Scholar]
- Casper C. Valganciclovir for suppression of human herpesvirus-8 replication: a randomized, double-blind, placebo-controlled, crossover trial. J Infect Dis. 2008;198:23–30. doi: 10.1086/588820. (Ref.189) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Centers for Disease Control and Prevention (CDC). Imported Lassa fever—New Jersey, 2004. MMWR Morb Mortal Wkly Rep. 2004;53:894–897. (Ref.27) [PubMed] [Google Scholar]
- Centers for Disease Control and Prevention (CDC). Rift Valley fever outbreaks—Kenya November 2006-January 2007. MMWR Morb Mortal Wkly Rep. 2007;56:73–76. (Ref.52) [PubMed] [Google Scholar]
- Centers for Disease Control and Prevention (CDC). Update: chikungunya fever diagnosed among international travelers—United States, 2006. MMWR Morb Mortal Wkly Rep. 2007;56:276–277. (Ref.3) [PubMed] [Google Scholar]
- Chan HL-Y. Clinical significance of hepatic derangement in severe acute respiratory syndrome. World J Gastroenterol. 2005;11:2148–2153. doi: 10.3748/wjg.v11.i14.2148. (Ref.215) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang YL. Human herpesvirus 6–related fulminant myocarditis in an immunocompetent adult with fatal outcome. Hum Pathol. 2009;40:740–745. doi: 10.1016/j.humpath.2008.08.017. (Ref.149) [DOI] [PubMed] [Google Scholar]
- Charrel RN, de Lamballerie X, Raoult D. Chikungunya outbreaks—the globalization of vector-borne diseases. N Engl J Med. 2007;356:769–771. doi: 10.1056/NEJMp078013. (Ref.71) [DOI] [PubMed] [Google Scholar]
- Chau TN. SARS-associated viral hepatitis caused by a novel coronavirus: report of three cases. Hepatology. 2004;39:302–310. doi: 10.1002/hep.20111. (Ref.216) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chevret L. Human herpesvirus-6 infection: a prospective study evaluating HHV-6 DNA levels in liver from children with acute liver failure. J Med Virol. 2008;80:1051–1057. doi: 10.1002/jmv.21143. (Ref.146) [DOI] [PubMed] [Google Scholar]
- Choi KW. Outcomes and prognostic factors in 267 patients with severe acute respiratory syndrome in Hong Kong. Ann Intern Med. 2003;139:715–723. doi: 10.7326/0003-4819-139-9-200311040-00005. (Ref.213) [DOI] [PubMed] [Google Scholar]
- Choquet S. Efficacy and safety of rituximab in B-cell post-transplantation lymphoproliferative disorders: results of a prospective multicentre phase 2 study. Blood. 2006;107:3053–3057. doi: 10.1182/blood-2005-01-0377. (Ref.122) [DOI] [PubMed] [Google Scholar]
- Colebunders R. Marburg hemorrhagic fever in Durba and Watsa, Democratic Republic of the Congo: clinical documentation, features of illness, and treatment. J Infect Dis. 2007;196(Suppl 2):S148–S153. doi: 10.1086/520543. (Ref.40) [DOI] [PubMed] [Google Scholar]
- Deback C. Detection of human herpesviruses HHV-6, HHV-7 and HHV-8 in whole blood by real-time PCR using the new CMV, HHV-6, 7, 8 R-gene kit. J Virol Methods. 2008;149:285–291. doi: 10.1016/j.jviromet.2008.01.026. (Ref.163) [DOI] [PubMed] [Google Scholar]
- Di Trolio R. Role of pegylated liposomal doxorubicin (PLD) in systemic Kaposi's sarcoma: a systematic review. Int J Immunopathol Pharmacol. 2006;19:253–263. doi: 10.1177/039463200601900202. (Ref.186) [DOI] [PubMed] [Google Scholar]
- Drosten C. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N Engl J Med. 2003;348:1967–1976. doi: 10.1056/NEJMoa030747. (Ref.209) [DOI] [PubMed] [Google Scholar]
- Drosten C. Molecular diagnostics of viral haemorrhagic fevers. Antiviral Res. 2003;57:61–87. doi: 10.1016/s0166-3542(02)00201-2. (Ref.54) [DOI] [PubMed] [Google Scholar]
- Eisenhut M. Ischemic hepatitis and collateral damage to the liver in severe viral respiratory tract infections. Am J Pathol. 2006;169:1100. doi: 10.2353/ajpath.2006.060514. (Ref.207) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eisenhut M, Thorburn K, Ahmed T. Transaminase levels in ventilated children with respiratory syncytial virus bronchiolitis. Intensive Care Med. 2004;30:931–934. doi: 10.1007/s00134-004-2236-2. (Ref.206) [DOI] [PubMed] [Google Scholar]
- Engelmann I. Rapid quantitative PCR assay for the simultaneous detection of herpes simplex virus, varicella zoster virus, cytomegalovirus, Epstein-Barr virus, and human herpesvirus 6 DNA in blood and other clinical specimens. J Med Virol. 2008;80:467–477. doi: 10.1002/jmv.21095. (Ref.164) [DOI] [PubMed] [Google Scholar]
- Engles EA. Risk factor for human herpesvirus 8 infection among adults in the United States and evidence for sexual transmission. J Infect Dis. 2007;196:199–207. doi: 10.1086/518791. (Ref.175) [DOI] [PubMed] [Google Scholar]
- Enria DA, Briggiler AM, Sánchez Z. Treatment of Argentine hemorrhagic fever. Antiviral Res. 2008;78:132–139. doi: 10.1016/j.antiviral.2007.10.010. (Ref.35) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ergönül Ö. Crimean-Congo haemorrhagic fever. Lancet Infect Dis. 2006;6:203–214. doi: 10.1016/S1473-3099(06)70435-2. (Ref.53) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ergönül Ö. Analysis of the mortality among the patients with Crimean Congo hemorrhagic fever virus infection: severity criteria revisited. Clin Microbiol. 2006;12:551–554. doi: 10.1111/j.1469-0691.2006.01445.x. (Ref.56) [DOI] [PubMed] [Google Scholar]
- Ergönül Ö. The characteristics of Crimean-Congo hemorrhagic fever in a recent outbreak in Turkey and the impact of oral ribavirin therapy. Clin Infect Dis. 2004;39:285–289. doi: 10.1086/422000. (Ref.55) [DOI] [PubMed] [Google Scholar]
- Finström N. Analysis of varicella-zoster virus and herpes simplex virus in various clinical samples by the use of different PCR assays. J Virol Methods. 2009;150:193–196. doi: 10.1016/j.jviromet.2009.04.035. (Ref.93) [DOI] [PubMed] [Google Scholar]
- Flamand L. Multicenter comparison of PCR assays for detection of human herpesvirus 6 DNA in serum. J Clin Microbiol. 2008;46:2700–2706. doi: 10.1128/JCM.00370-08. (Ref.165) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fohrer C. Long-term survival in post-transplant lymphoproliferative disorders with a dose-adjusted ACVBP regimen. Br J Haematol. 2006;134:602–612. doi: 10.1111/j.1365-2141.2006.06228.x. (Ref.119) [DOI] [PubMed] [Google Scholar]
- Frederick DM, Bland D, Gollin Y. Fatal disseminated herpes simplex virus infection in a previously healthy pregnant woman. J Reprod Med. 2002;47:591–596. (Ref.90) [PubMed] [Google Scholar]
- Freeman DO. Spectrum of disease and relation to place of exposure among ill returned travelers. N Engl J Med. 2006;354:119–130. doi: 10.1056/NEJMoa051331. (Ref.1) [DOI] [PubMed] [Google Scholar]
- Garnett CT. Latent species C adenoviruses in human tonsil tissues. J Virol. 2009;83:2417–2428. doi: 10.1128/JVI.02392-08. (Ref.191) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gross TG. Low dose chemotherapy of Epstein-Barr virus–positive post-transplantation lymphoproliferative disease in children after solid organ transplantation. J Clin Oncol. 2005;23:6481–6488. doi: 10.1200/JCO.2005.08.074. (Ref.118) [DOI] [PubMed] [Google Scholar]
- Haas WH. Imported Lassa fever in Germany: surveillance and management of contact persons. Clin Infect Dis. 2003;36:1254–1258. doi: 10.1086/374853. (Ref.26) [DOI] [PubMed] [Google Scholar]
- Härmä M. Pre-transplant human herpesvirus 6 infection of patients with acute liver failure is a risk factor for post transplant human herpesvirus 6 infection of the liver. Transplantation. 2006;81:367–372. doi: 10.1097/01.tp.0000195771.83614.0b. (Ref.154) [DOI] [PubMed] [Google Scholar]
- Härmä M, Höckerstedt K, Lautenschlager I. Human herpesvirus-6 and acute liver failure. Transplantation. 2003;76:536–539. doi: 10.1097/01.TP.0000069233.13409.DF. (Ref.147) [DOI] [PubMed] [Google Scholar]
- Hayakawa H. A clinical study of adult human parvovirus B 19 infection. Intern Med. 2007;41:295–299. doi: 10.2169/internalmedicine.41.295. (Ref.239) [DOI] [PubMed] [Google Scholar]
- Hsiao CH. Immunohistochemical study of severe acute respiratory syndrome–associated coronavirus in tissue sections of patients. J Formos Med Assoc. 2005;104:150–156. (Ref.218) [PubMed] [Google Scholar]
- Isegawa Y. Human herpesvirus 6 ganciclovir-resistant strain with amino acid substitutions associated with the death of an allogeneic stem cell transplant recipient. J Clin Virol. 2009;44:15–19. doi: 10.1016/j.jcv.2008.09.002. (Ref.167) [DOI] [PubMed] [Google Scholar]
- Jessie K. Localization of dengue virus in naturally infected human tissues, by immunohistochemistry and in situ hybridization. J Infect Dis. 2004;189:1411–1418. doi: 10.1086/383043. (Ref.7) [DOI] [PubMed] [Google Scholar]
- Kojaoghlanian T, Flomenberg P, Horwitz MS. The impact of adenovirus infection on the immunocompromised host. Rev Med Virol. 2003;13:155–171. doi: 10.1002/rmv.386. (Ref.192) [DOI] [PubMed] [Google Scholar]
- Koreishi A. Synchronous follicular lymphoma, Kaposi sarcoma, and Castleman's disease in a HIV-negative patient with EBV and HHV-8 coinfection. Int J Surg Pathol. 2009 doi: 10.1177/1066896909341803. Aug 5 [Epub ahead of print].(Ref.180) [DOI] [PubMed] [Google Scholar]
- Ksiazek TG. A novel coronavirus in patients with severe acute respiratory syndrome. N Engl J Med. 2003;348:1953–1966. doi: 10.1056/NEJMoa030781. (Ref.208) [DOI] [PubMed] [Google Scholar]
- Kuntzen T. Postinfantile giant cell hepatitis with autoimmune features following a human herpesvirus 6–induced adverse drug reaction. Eur J Gastroenterol Hepatol. 2005;17:1131–1134. doi: 10.1097/00042737-200510000-00020. (Ref.160) [DOI] [PubMed] [Google Scholar]
- Kylat RI, Kelly EN, Ford-Jones EL. Clinical findings and adverse outcome in neonates with symptomatic congenital cytomegalovirus (SCCMV) infection. Eur J Pediatr. 2006;165:773–778. doi: 10.1007/s00431-006-0172-6. (Ref.126) [DOI] [PubMed] [Google Scholar]
- Lee N. A major outbreak of severe acute respiratory syndrome in Hong Kong. N Engl J Med. 2003;348:1986–1994. doi: 10.1056/NEJMoa030685. (Ref.210) [DOI] [PubMed] [Google Scholar]
- Lee WM. Brief report: no evidence for parvovirus B19 or hepatitis E as a cause of acute liver failure. Dig Dis Sci. 2006;51:1712–1715. doi: 10.1007/s10620-005-9061-5. (Ref.244) [DOI] [PubMed] [Google Scholar]
- Leroy EM. Fruit bats as reservoir of Ebola virus. Nature. 2005;438:575–576. doi: 10.1038/438575a. (Ref.39) [DOI] [PubMed] [Google Scholar]
- Levin MJ, Bacon TH, Leary JJ. Resistance of herpes simplex virus infections to nucleoside analogues in HIV-infected patients. Clin Infect Dis. 2004;39(Suppl 5):S248–S257. doi: 10.1086/422364. (Ref.98) [DOI] [PubMed] [Google Scholar]
- Lledó L. Hantavirus infections in Spain: analysis of sera from the general population and from patients with pneumonia, renal disease and hepatitis. J Clin Virol. 2003;27:296–307. doi: 10.1016/s1386-6532(02)00228-7. (Ref.66) [DOI] [PubMed] [Google Scholar]
- Lo AW, Tang NLS, To KF. How the SARS coronavirus causes disease: host or organism? J Pathol. 2006;208:142–151. doi: 10.1002/path.1897. (Ref.219) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahanty S, Bray M. Pathogenesis of filoviral haemorrhagic fevers. Lancet Infect Dis. 2004;4:487–498. doi: 10.1016/S1473-3099(04)01103-X. (Ref.37) [DOI] [PubMed] [Google Scholar]
- Mardani M. The efficacy of oral ribavirin in the treatment of Crimean-Congo hemorrhagic fever in Iran. Clin Infect Dis. 2003;36:1613–1618. doi: 10.1086/375058. (Ref.61) [DOI] [PubMed] [Google Scholar]
- Martin BK. Change over time in incidence of ganciclovir resistance in patients with cytomegalovirus retinitis. Clin Infect Dis. 2008;44:1001–1008. doi: 10.1086/512368. (Ref.140) [DOI] [PubMed] [Google Scholar]
- Nakazibwe C. Marburg virus outbreak leads scientists to suspected disease reservoir. Bull World Health Organ. 2007;85:654–656. doi: 10.2471/BLT.07.020907. (Ref.41) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Naresh KN, Rice AJ, Bower M. Lymph nodes involved by multicentric Castleman disease among HIV-positive individuals are often involved by Kaposi sarcoma. Am J Surg Pathol. 2008;32:1006–1012. doi: 10.1097/PAS.0b013e318160ed97. (Ref.179) [DOI] [PubMed] [Google Scholar]
- Nobili V. Acute liver failure as presenting feature in tyrosinemia type 1 in a child with primary HHV-6 infection. J Gastroenterol Hepatol. 2006;21:339. doi: 10.1111/j.1440-1746.2006.04045.x. (Ref.148) [DOI] [PubMed] [Google Scholar]
- Oertel SH. Effect of anti-CD20 antibody rituximab in patients with post-transplant lymphoproliferative disorder (PTLD) Am J Transplant. 2005;5:2901–2906. doi: 10.1111/j.1600-6143.2005.01098.x. (Ref.121) [DOI] [PubMed] [Google Scholar]
- Ogbu O, Ajuluchukwu E, Uneke CJ. Lassa fever in West African subregion: an overview. J Vector Borne Dis. 2007;44:1–11. (Ref.28) [PubMed] [Google Scholar]
- Ohashi M. Human herpesvirus 6 infection in adult living related liver transplant recipients. Liver Transpl. 2008;14:100–109. doi: 10.1002/lt.21304. (Ref.155) [DOI] [PubMed] [Google Scholar]
- Peiris JS. Re-emergence of fatal human influenza A subtype H5N1 disease. Lancet. 2004;363:617–619. doi: 10.1016/S0140-6736(04)15595-5. (Ref.205) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peters CJ. Marburg and Ebola—arming ourselves against the deadly filoviruses. N Engl J Med. 2005;352:2571–2573. doi: 10.1056/NEJMp058109. (Ref.36) [DOI] [PubMed] [Google Scholar]
- Piaggio F. Torque Teno virus—cause of viral liver disease following liver transplantation: a case report. Transplant Proc. 2009;41:1378–1379. doi: 10.1016/j.transproceed.2009.03.047. (Ref.250) [DOI] [PubMed] [Google Scholar]
- Pinna AD. Five cases of fulminant hepatitis due to herpes simplex virus in adults. Dig Dis Sci. 2002;47:750–754. doi: 10.1023/a:1014779614525. (Ref.83) [DOI] [PubMed] [Google Scholar]
- Piselli P. Risk of Kaposi sarcoma after solid-organ transplantation: multicenter study in 4,767 recipients in Italy, 1970-2006. Transplant Proc. 2009;41:1227–1230. doi: 10.1016/j.transproceed.2009.03.009. (Ref.176) [DOI] [PubMed] [Google Scholar]
- Polakos NK. Kupffer cell–dependent hepatitis occurs during influenza infection. Am J Pathol. 2006;168:1169–1178. doi: 10.2353/ajpath.2006.050875. (Ref.200) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Potenza I. HHV-6A in syncytial giant-cell hepatitis. N Engl J Med. 2008;359:583–602. doi: 10.1056/NEJMoa074479. (Ref.159) [DOI] [PubMed] [Google Scholar]
- Pourrut X. Spatial and temporal patterns of Zaire Ebola virus antibody prevalence in the possible reservoir bat species. J Infect Dis. 2007;196(Suppl 2):S176–S183. doi: 10.1086/520541. (Ref.42) [DOI] [PubMed] [Google Scholar]
- Poutanen SM. Identification of severe acute respiratory syndrome in Canada. N Engl J Med. 2003;348:1995–2005. doi: 10.1056/NEJMoa030634. (Ref.212) [DOI] [PubMed] [Google Scholar]
- Quaresma JA. Reconsiderations of histopathology and ultrastructural aspects of the human liver in yellow fever. Acta Trop. 2005;94:116–127. doi: 10.1016/j.actatropica.2005.03.003. (Ref.17) [DOI] [PubMed] [Google Scholar]
- Quaresma JA. Immunohistochemical examination of the role of Fas ligand and lymphocytes in the pathogenesis of human liver yellow fever. Virus Res. 2006;116:91–97. doi: 10.1016/j.virusres.2005.08.019. (Ref.20) [DOI] [PubMed] [Google Scholar]
- Quaresma JA. Revisiting the liver in human yellow fever: virus-induced apoptosis in hepatocytes associated with TGF-beta, TNF-alpha and NK cell activity. Virology. 2006;345:22–30. doi: 10.1016/j.virol.2005.09.058. (Ref.19) [DOI] [PubMed] [Google Scholar]
- Rezza G. Infection with chikungunya virus in Italy: an outbreak in a temperate region. Lancet. 2007;370:1840–1846. doi: 10.1016/S0140-6736(07)61779-6. (Ref.72) [DOI] [PubMed] [Google Scholar]
- Shi X. Severe acute respiratory syndrome–associated coronavirus is detected in intestinal D tissues of fatal cases. Am J Gastroenterol. 2005;100:169–176. doi: 10.1111/j.1572-0241.2005.40377.x. (Ref.217) [DOI] [PubMed] [Google Scholar]
- Sidwell RW, Smee DF. Viruses of the Bunya- and Togaviridae families: potential as bioterrorism agents and means of control. Antiviral Res. 2003;57:101–111. doi: 10.1016/s0166-3542(02)00203-6. (Ref.70) [DOI] [PubMed] [Google Scholar]
- Spengler U. Hepatic toxicity of antiviral agents. In: Kaplowith N, De Leve LD, editors. Drug induced liver disease. Informa Health Care USA; New York: 2007. pp. 567–591. (Ref.82) [Google Scholar]
- Stebbing J. Paclitaxel for anthracycline-resistant AIDS-related Kaposi's sarcoma: clinical and angiogenic correlations. Ann Oncol. 2003;14:1660–1666. doi: 10.1093/annonc/mdg461. (Ref.187) [DOI] [PubMed] [Google Scholar]
- Tasdelen NF. The role of ribavirin in the therapy of Crimean-Congo hemorrhagic fever: early use is promising. Eur J Clin Microbiol Infect Dis. 2009;28:929–933. doi: 10.1007/s10096-009-0728-2. (Ref.62) [DOI] [PubMed] [Google Scholar]
- Taubitz W. Chikungunya fever in travelers: clinical presentation and course. Clin Infect Dis. 2007;45:e1–e4. doi: 10.1086/518701. (Ref.73) [DOI] [PubMed] [Google Scholar]
- Towner JS. Marburg virus infection detected in a common African bat. PLoS One. 2007;2:e764. doi: 10.1371/journal.pone.0000764. (Ref.43) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van der Ende M. Complete clinical and virological remission of refractory HIV-related Kaposi's sarcoma with pegylated interferon alpha. AIDS. 2007;21:1661–1662. doi: 10.1097/QAD.0b013e32827035a4. (Ref.185) [DOI] [PubMed] [Google Scholar]
- Vasconcelos PF. Genetic divergence and dispersal of yellow fever virus, Brazil. Emerg Infect Dis. 2004;10:1578–1584. doi: 10.3201/eid1009.040197. (Ref.14) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waza K, Inoue K, Matsumura S. Symptoms associated with parvovirus B19 infection in adults: a pilot study. Intern Med. 2007;46:1975–1978. doi: 10.2169/internalmedicine.46.0366. (Ref.240) [DOI] [PubMed] [Google Scholar]
- Weinberger B. Quantitation of Epstein-Barr virus mRNA using reverse transcription and real-time PCR. J Med Virol. 2004;74:612–618. doi: 10.1002/jmv.20220. (Ref.116) [DOI] [PubMed] [Google Scholar]
- Wilder-Smith A, Schwartz E. Dengue in travelers. N Engl J Med. 2005;353:924–932. doi: 10.1056/NEJMra041927. (Ref.4) [DOI] [PubMed] [Google Scholar]
- Wilson ME. Fever in returned travelers: results from the GeoSentinel Surveillance Network. Clin Infect Dis. 2007;44:1560–1568. doi: 10.1086/518173. (Ref.2) [DOI] [PubMed] [Google Scholar]
- Wong WM. Temporal pattern of hepatic dysfunction and disease severity in patients with SARS. JAMA. 2003;290:2663–2665. doi: 10.1001/jama.290.20.2663. (Ref.214) [DOI] [PubMed] [Google Scholar]
References
- 1.Freeman DO. Spectrum of disease and relation to place of exposure among ill returned travelers. N Engl J Med. 2006;354:119–130. doi: 10.1056/NEJMoa051331. [DOI] [PubMed] [Google Scholar]
- 2.Wilson ME. Fever in returned travelers: results from the GeoSentinel Surveillance Network. Clin Infect Dis. 2007;44:1560–1568. doi: 10.1086/518173. [DOI] [PubMed] [Google Scholar]
- 3.Centers for Disease Control and Prevention (CDC) Update: chikungunya fever diagnosed among international travelers—United States, 2006. MMWR Morb Mortal Wkly Rep. 2007;56:276–277. [PubMed] [Google Scholar]
- 4.Wilder-Smith A, Schwartz E. Dengue in travelers. N Engl J Med. 2005;353:924–932. doi: 10.1056/NEJMra041927. [DOI] [PubMed] [Google Scholar]
- 5.Rosen L, Khin MM, U T. Recovery of virus from the liver of children with fatal dengue: reflections on the pathogenesis of the disease and its possible analogy with that of yellow fever. Res Virol. 1989;140:351–360. doi: 10.1016/s0923-2516(89)80115-3. [DOI] [PubMed] [Google Scholar]
- 6.Hall WC. Demonstration of yellow fever and dengue antigens in formalin-fixed paraffin-embedded human liver by immunohistochemical analysis. Am J Trop Med Hyg. 1991;45:408–417. doi: 10.4269/ajtmh.1991.45.408. [DOI] [PubMed] [Google Scholar]
- 7.Jessie K. Localization of dengue virus in naturally infected human tissues, by immunohistochemistry and in situ hybridization. J Infect Dis. 2004;189:1411–1418. doi: 10.1086/383043. [DOI] [PubMed] [Google Scholar]
- 8.Kalayanarooj S. Early clinical and laboratory indicators of acute dengue illness. J Infect Dis. 1997;176:313–321. doi: 10.1086/514047. [DOI] [PubMed] [Google Scholar]
- 9.Wahid SF. A comparison of the pattern of liver involvement in dengue hemorrhagic fever with classic dengue fever. Southeast Asian J Trop Med Public Health. 2000;31:259–263. [PubMed] [Google Scholar]
- 10.Bhamarapravati N, Tuchinda P, Booyapaknavik V. Pathology of Thailand haemorrhagic fever: a study of 100 autopsy cases. Ann Trop Med Parasitol. 1967;61:500–510. doi: 10.1080/00034983.1967.11686519. [DOI] [PubMed] [Google Scholar]
- 11.Huerre MR. Liver histopathology and biological correlates in five cases of fatal dengue fever in Vietnamese children. Virchows Arch. 2001;438:107–115. doi: 10.1007/s004280000329. [DOI] [PubMed] [Google Scholar]
- 12.Marianneau P. Dengue virus replication in human hepatoma cells activates NF-κB which in turn induces apoptotic cell death. J Virol. 1997;71:3244–3249. doi: 10.1128/jvi.71.4.3244-3249.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Mutebi JP. Phylogenetic and evolutionary relationships among yellow fever virus isolates in Africa. J Virol. 2001;75:6999–7008. doi: 10.1128/JVI.75.15.6999-7008.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Vasconcelos PF. Genetic divergence and dispersal of yellow fever virus, Brazil. Emerg Infect Dis. 2004;10:1578–1584. doi: 10.3201/eid1009.040197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Barnett ED. Yellow fever: epidemiology and prevention. Clin Infect Dis. 2007;44:850–856. doi: 10.1086/511869. [DOI] [PubMed] [Google Scholar]
- 16.Oudart JL, Rey M. [Proteinuria, proteinaemia, and serum transaminase activity in 23 confirmed cases of yellow fever.] Bull World Health Organ. 1970;43:95–102. [PMC free article] [PubMed] [Google Scholar]
- 17.Quaresma JA. Reconsiderations of histopathology and ultrastructural aspects of the human liver in yellow fever. Acta Trop. 2005;94:116–127. doi: 10.1016/j.actatropica.2005.03.003. [DOI] [PubMed] [Google Scholar]
- 18.Bae HG. Analysis of two imported cases of yellow fever infection from Ivory Coast and The Gambia to Germany and Belgium. J Clin Virol. 2005;33:274–280. doi: 10.1016/j.jcv.2004.12.001. [DOI] [PubMed] [Google Scholar]
- 19.Quaresma JA. Revisiting the liver in human yellow fever: virus-induced apoptosis in hepatocytes associated with TGF-beta, TNF-alpha and NK cell activity. Virology. 2006;345:22–30. doi: 10.1016/j.virol.2005.09.058. [DOI] [PubMed] [Google Scholar]
- 20.Quaresma JA. Immunohistochemical examination of the role of Fas ligand and lymphocytes in the pathogenesis of human liver yellow fever. Virus Res. 2006;116:91–97. doi: 10.1016/j.virusres.2005.08.019. [DOI] [PubMed] [Google Scholar]
- 21.Francis TL. A clinicopathological study of human yellow fever. Bull World Health Organ. 1972;46:659–667. [PMC free article] [PubMed] [Google Scholar]
- 22.Gabrielsen B. Antiviral (RNA) activity of selected Amaryllidaceae isoquinoline constituents and synthesis of related substances. J Nat Prod. 1992;55:1569–1581. doi: 10.1021/np50089a003. [DOI] [PubMed] [Google Scholar]
- 23.Monath TP. Lassa virus isolation from Mastomys natalensis rodents during an epidemic in Sierra Leone. Science. 1974;185:263–265. doi: 10.1126/science.185.4147.263. [DOI] [PubMed] [Google Scholar]
- 24.Carey DE. Lassa fever. Epidemiological aspects of the 1970 epidemic, Jos, Nigeria. Trans R Soc Trop Med Hyg. 1972;66:402–408. doi: 10.1016/0035-9203(72)90271-4. [DOI] [PubMed] [Google Scholar]
- 25.Johnson KM. Lassa fever in the United States: investigation of a case and new guidelines for management. N Engl J Med. 1990;323:1120–1123. doi: 10.1056/NEJM199010183231607. [DOI] [PubMed] [Google Scholar]
- 26.Haas WH. Imported Lassa fever in Germany: surveillance and management of contact persons. Clin Infect Dis. 2003;36:1254–1258. doi: 10.1086/374853. [DOI] [PubMed] [Google Scholar]
- 27.Centers for Disease Control and Prevention (CDC) Imported Lassa fever—New Jersey, 2004. MMWR Morb Mortal Wkly Rep. 2004;53:894–897. [PubMed] [Google Scholar]
- 28.Ogbu O, Ajuluchukwu E, Uneke CJ. Lassa fever in West African subregion: an overview. J Vector Borne Dis. 2007;44:1–11. [PubMed] [Google Scholar]
- 29.Bausch DG. Lassa fever in Guinea: I. Epidemiology of human disease and clinical observations. Vector Borne Zoonotic Dis. 2001;1:269–281. doi: 10.1089/15303660160025903. [DOI] [PubMed] [Google Scholar]
- 30.Jahrling PB, Niklasson BS, McCormick JB. Early diagnosis of human Lassa fever by ELISA detection of antigen and antibody. Lancet. 1985;1:250–252. doi: 10.1016/s0140-6736(85)91029-3. [DOI] [PubMed] [Google Scholar]
- 31.Trappier SG. Evaluation of the polymerase chain reaction for diagnosis of Lassa virus infection. Am J Trop Med Hyg. 1993;49:214–221. doi: 10.4269/ajtmh.1993.49.214. [DOI] [PubMed] [Google Scholar]
- 32.Jahrling PB. Lassa virus infection of rhesus monkeys: pathogenesis and treatment with ribavirin. J infect Dis. 1980;141:580–589. doi: 10.1093/infdis/141.5.580. [DOI] [PubMed] [Google Scholar]
- 33.McCormick JB. Lassa fever: effective therapy with ribavirin. N Engl J Med. 1986;314:20–26. doi: 10.1056/NEJM198601023140104. [DOI] [PubMed] [Google Scholar]
- 34.Harrison LH. Clinical case definitions for Argentine hemorrhagic fever. Clin Infect Dis. 1999;28:1091–1094. doi: 10.1086/514749. [DOI] [PubMed] [Google Scholar]
- 35.Enria DA, Briggiler AM, Sánchez Z. Treatment of Argentine hemorrhagic fever. Antiviral Res. 2008;78:132–139. doi: 10.1016/j.antiviral.2007.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Peters CJ. Marburg and Ebola—arming ourselves against the deadly filoviruses. N Engl J Med. 2005;352:2571–2573. doi: 10.1056/NEJMp058109. [DOI] [PubMed] [Google Scholar]
- 37.Mahanty S, Bray M. Pathogenesis of filoviral haemorrhagic fevers. Lancet Infect Dis. 2004;4:487–498. doi: 10.1016/S1473-3099(04)01103-X. [DOI] [PubMed] [Google Scholar]
- 38.Alsop Z. Ebola outbreak in Uganda “atypical,” say experts. Lancet. 2007;370:2085. doi: 10.1016/s0140-6736(07)61880-7. [DOI] [PubMed] [Google Scholar]
- 39.Leroy EM. Fruit bats as reservoir of Ebola virus. Nature. 2005;438:575–576. doi: 10.1038/438575a. [DOI] [PubMed] [Google Scholar]
- 40.Colebunders R. Marburg hemorrhagic fever in Durba and Watsa, Democratic Republic of the Congo: clinical documentation, features of illness, and treatment. J Infect Dis. 2007;196(Suppl 2):S148–S153. doi: 10.1086/520543. [DOI] [PubMed] [Google Scholar]
- 41.Nakazibwe C. Marburg virus outbreak leads scientists to suspected disease reservoir. Bull World Health Organ. 2007;85:654–656. doi: 10.2471/BLT.07.020907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Pourrut X. Spatial and temporal patterns of Zaire Ebola virus antibody prevalence in the possible reservoir bat species. J Infect Dis. 2007;196(Suppl 2):S176–S183. doi: 10.1086/520541. [DOI] [PubMed] [Google Scholar]
- 43.Towner JS. Marburg virus infection detected in a common African bat. PLoS One. 2007;2:e764. doi: 10.1371/journal.pone.0000764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Peters CJ, Jahrling PB, Khan AS. Patients infected with high-hazard viruses: scientific basis for infection control. Arch Virol. 1996;11:141–168. doi: 10.1007/978-3-7091-7482-1_13. [DOI] [PubMed] [Google Scholar]
- 45.Bwaka MA. Ebola hemorrhagic fever in Kikwit, Democratic Republic of the Congo: clinical observations in 103 patients. J Infect Dis. 1999;179(Suppl 1):S1–S7. doi: 10.1086/514308. [DOI] [PubMed] [Google Scholar]
- 46.Formenty P. Human infection due to Ebola virus, subtype Cote d’Ivoire: clinical and biological presentation. J Infect Dis. 1999;179(Suppl 1):S48–S53. doi: 10.1086/514285. [DOI] [PubMed] [Google Scholar]
- 47.Bechtelsheimer H, Korb G, Gedigk P. The morphology and pathogenesis of “Marburg Virus” hepatitis. Hum Pathol. 1972;3:255–263. doi: 10.1016/s0046-8177(72)80079-0. [DOI] [PubMed] [Google Scholar]
- 48.Rippey JJ, Schepers NJ, Gear JHS. The pathology of Marburg virus disease. S Afr Med J. 1984;66:50–54. [PubMed] [Google Scholar]
- 49.Zaki SR. Ebola virus hemorrhagic fever: pathologic, immunopathologic and ultrastructural studies. Lab Invest. 1996;74:133A. [Google Scholar]
- 50.Rowe AK. Clinical, virologic, and immunologic follow-up of convalescent Ebola hemorrhagic fever patients and their household contacts, Kikwit, Democratic Republic of the Congo. Commission de Lutte contre les Epidémies à Kikwit. J Infect Dis. 1999;179(Suppl 1):S28–S35. doi: 10.1086/514318. [DOI] [PubMed] [Google Scholar]
- 51.Daubney R, Hudson JR, Garnham PC. Enzootic hepatitis or Rift Valley fever: an undescribed virus disease of sheep cattle and man from East Africa. J Pathol Bacteriol. 1931;35:545–579. [Google Scholar]
- 52.Centers for Disease Control and Prevention (CDC) Rift Valley fever outbreaks—Kenya November 2006-January 2007. MMWR Morb Mortal Wkly Rep. 2007;56:73–76. [PubMed] [Google Scholar]
- 53.Ergönül Ö. Crimeasn-Congo haemorrhagic fever. Lancet Infect Dis. 2006;6:203–214. doi: 10.1016/S1473-3099(06)70435-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Drosten C. Molecular diagnostics of viral haemorrhagic fevers. Antiviral Res. 2003;57:61–87. doi: 10.1016/s0166-3542(02)00201-2. [DOI] [PubMed] [Google Scholar]
- 55.Ergönül Ö. The characteristics of Crimean-Congo hemorrhagic fever in a recent outbreak in Turkey and the impact of oral ribavirin therapy. Clin Infect Dis. 2004;39:285–289. doi: 10.1086/422000. [DOI] [PubMed] [Google Scholar]
- 56.Ergönül Ö. Analysis of the mortality among the patients with Crimean Congo hemorrhagic fever virus infection: severity criteria revisited. Clin Microbiol. 2006;12:551–554. doi: 10.1111/j.1469-0691.2006.01445.x. [DOI] [PubMed] [Google Scholar]
- 57.Burt FJ. Immunohistochemical and in situ localization of Crimean-Congo hemorrhagic fever (CCHF) virus in human tissues and implications for CCHF pathogenesis. Arch Pathol Lab Med. 1997;121:839–846. [PubMed] [Google Scholar]
- 58.Watts DM. Inhibition of Crimean-Congo hemorrhagic fever viral infectivity yields in vitro by ribavirin. Am J Trop Med Hyg. 1989;41:581–585. doi: 10.4269/ajtmh.1989.41.581. [DOI] [PubMed] [Google Scholar]
- 59.Tignor GH, Hanham CA. Ribavirin efficacy in an in vivo model of Crimean-Congo hemorrhagic fever virus (CCHF) infection. Antiviral Res. 1993;22:309–325. doi: 10.1016/0166-3542(93)90040-P. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Fischer-Hoch SP. Crimean-Congo hemorrhagic fever treated with oral ribavirin. Lancet. 1995;346:472–475. doi: 10.1016/s0140-6736(95)91323-8. [DOI] [PubMed] [Google Scholar]
- 61.Mardani M. The efficacy of oral ribavirin in the treatment of Crimean-Congo hemorrhagic fever in Iran. Clin Infect Dis. 2003;36:1613–1618. doi: 10.1086/375058. [DOI] [PubMed] [Google Scholar]
- 62.Tasdelen NF. The role of ribavirin in the therapy of Crimean-Congo hemorrhagic fever: early use is promising. Eur J Clin Microbiol Infect Dis. 2009;28:929–933. doi: 10.1007/s10096-009-0728-2. [DOI] [PubMed] [Google Scholar]
- 63.Nichol TS, Aikawa J, Kawacka Y. Emerging viral diseases. Proc Natl Acad Sci U S A. 2000;97:12411–12412. doi: 10.1073/pnas.210382297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Plyusnin A, Krüger DH, Lundkvist A. Hantavirus infections in Europe. Adv Virus Res. 2001;57:105–136. doi: 10.1016/s0065-3527(01)57002-5. [DOI] [PubMed] [Google Scholar]
- 65.Chan YC. Haemorrhagic fever with renal syndrome involving the liver. Med J Aust. 1987;147:248–249. doi: 10.5694/j.1326-5377.1987.tb133422.x. [DOI] [PubMed] [Google Scholar]
- 66.Lledó L. Hantavirus infections in Spain: analysis of sera from the general population and from patients with pneumonia, renal disease and hepatitis. J Clin Virol. 2003;27:296–307. doi: 10.1016/s1386-6532(02)00228-7. [DOI] [PubMed] [Google Scholar]
- 67.Meng G. High prevalence of hantavirus infection in a group of Chinese patients with acute hepatitis of unknown aetiology. J Viral Hepat. 1997;4:231–234. doi: 10.1046/j.1365-2893.1997.00140.x. [DOI] [PubMed] [Google Scholar]
- 68.Kariwa H. Detection of hantaviral antibodies among patients with hepatitis of unknown etiology in Japan. Microbiol Immunol. 2000;44:357–362. doi: 10.1111/j.1348-0421.2000.tb02506.x. [DOI] [PubMed] [Google Scholar]
- 69.Yotsuyanagi H. Acute exacerbation of autoimmune liver disease associated with hantaviral infection. Scand J Infect Dis. 1998;30:81–83. doi: 10.1080/003655498750002358. [DOI] [PubMed] [Google Scholar]
- 70.Sidwell RW, Smee DF. Viruses of the Bunya- and Togaviridae families: potential as bioterrorism agents and means of control. Antiviral Res. 2003;57:101–111. doi: 10.1016/s0166-3542(02)00203-6. [DOI] [PubMed] [Google Scholar]
- 71.Charrel RN, de Lamballerie X, Raoult D. Chikungunya outbreaks—the globalization of vector-borne diseases. N Engl J Med. 2007;356:769–771. doi: 10.1056/NEJMp078013. [DOI] [PubMed] [Google Scholar]
- 72.Rezza G. Infection with chikungunya virus in Italy: an outbreak in a temperate region. Lancet. 2007;370:1840–1846. doi: 10.1016/S0140-6736(07)61779-6. [DOI] [PubMed] [Google Scholar]
- 73.Taubitz W. Chikungunya fever in travelers: clinical presentation and course. Clin Infect Dis. 2007;45:e1–e4. doi: 10.1086/518701. [DOI] [PubMed] [Google Scholar]
- 74.Briolant S. In vitro inhibition of chikungunya and Semliki Forest viruses replication by antiviral compounds: synergistic effect of interferon-alpha and ribavirin combination. Antiviral Res. 2004;61:111–117. doi: 10.1016/j.antiviral.2003.09.005. [DOI] [PubMed] [Google Scholar]
- 75.Boag FC. Abnormalities of liver function during HIV seroconversion illness. Int J STD AIDS. 1992;3:46–48. doi: 10.1177/095646249200300111. [DOI] [PubMed] [Google Scholar]
- 76.Molina JM. Hepatitis associated with primary HIV infection. Gastroenterology. 1992;102:739. doi: 10.1016/0016-5085(92)90138-o. [DOI] [PubMed] [Google Scholar]
- 77.Welch K. Autopsy findings in the acquired immune deficiency syndrome. JAMA. 1984;252:1152–1159. [PubMed] [Google Scholar]
- 78.Freiman JP, Helfert KE, Hamrell MR, Stein DS. Hepatomegaly with severe steatosis in HIV-seropositive patients. AIDS. 1993;7:379–385. doi: 10.1097/00002030-199303000-00012. [DOI] [PubMed] [Google Scholar]
- 79.Housset C. Immunohistochemical evidence for human immunodeficiency virus-1 infection of the liver Kupffer cells. Hum Pathol. 1990;21:404–408. doi: 10.1016/0046-8177(90)90202-g. [DOI] [PubMed] [Google Scholar]
- 80.Cao Y. Identification and quantification of HIV-1 in the liver of patients with AIDS. AIDS. 1992;6:65–70. doi: 10.1097/00002030-199201000-00008. [DOI] [PubMed] [Google Scholar]
- 81.Lafon M-E KirnA. Human immunodeficiency virus infection of the liver. Semin Liver Dis. 1992;112:97–204. doi: 10.1055/s-2007-1007391. [DOI] [PubMed] [Google Scholar]
- 82.Spengler U. Hepatic toxicity of antiviral agents. In: Kaplowith N, De Leve LD, editors. Drug induced liver disease. Informa Health Care USA; New York: 2007. pp. 567–591. [Google Scholar]
- 83.Pinna AD. Five cases of fulminant hepatitis due to herpes simplex virus in adults. Dig Dis Sci. 2002;47:750–754. doi: 10.1023/a:1014779614525. [DOI] [PubMed] [Google Scholar]
- 84.Walker DP. Disseminated herpes simplex infection with hepatitis in an adult renal transplant recipient. J Clin Pathol. 1981;34:1044–1046. doi: 10.1136/jcp.34.9.1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Kusne S. Herpes simplex virus hepatitis after solid organ transplantation. J Infect Dis. 1991;163:1001–1007. doi: 10.1093/infdis/163.5.1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Johnson JR. Hepatitis due to herpes simplex virus in marrow-transplant recipients. Clin Infect Dis. 1992;14:38–45. doi: 10.1093/clinids/14.1.38. [DOI] [PubMed] [Google Scholar]
- 87.Kaufman B. Herpes simplex hepatitis: case report and review. Clin Infect Dis. 1997;24:334–338. doi: 10.1093/clinids/24.3.334. [DOI] [PubMed] [Google Scholar]
- 88.Zimmerli W. Disseminated herpes simplex type 2 and systemic Candida infection in a patient with previous asymptomatic HIV infection. J Infect Dis. 1988;157:597–598. doi: 10.1093/infdis/157.3.597. [DOI] [PubMed] [Google Scholar]
- 89.Hillard P, Seeds J, Cefalo R. Disseminated herpes simplex in pregnancy: two cases and a review. Obstet Gynecol Surv. 1982;37:449–453. doi: 10.1097/00006254-198207000-00002. [DOI] [PubMed] [Google Scholar]
- 90.Frederick DM, Bland D, Gollin Y. Fatal disseminated herpes simplex virus infection in a previously healthy pregnant woman. J Reprod Med. 2002;47:591–596. [PubMed] [Google Scholar]
- 91.Goodman ZD, Ishak KG, Sesterhenn IA. Herpes simplex hepatitis in apparently immunocompetent adults. Am J Clin Pathol. 1986;85:694–699. doi: 10.1093/ajcp/85.6.694. [DOI] [PubMed] [Google Scholar]
- 92.Bhattarakosol P, Yoosook C, Cross A. Intertypic variation of herpes simplex virus type 2 isolates detected by monoclonal antibodies against viral glycoproteins. Arch Virol. 1990;115:89–100. doi: 10.1007/BF01310625. [DOI] [PubMed] [Google Scholar]
- 93.Finström N. Analysis of varicella-zoster virus and herpes simplex virus in various clinical samples by the use of different PCR assays. J Virol Methods. 2009;150:193–196. doi: 10.1016/j.jviromet.2009.04.035. [DOI] [PubMed] [Google Scholar]
- 94.Vestey JP, Norval M. Mucocutaneous infections with herpes simplex virus and their management. Clin Exp Dermatol. 1992;17:221–237. doi: 10.1111/j.1365-2230.1992.tb02155.x. [DOI] [PubMed] [Google Scholar]
- 95.Seale L. Prevention of herpesvirus infections in renal allograft recipient. JAMA. 1985;254:3435–3438. [PubMed] [Google Scholar]
- 96.Pettersson E. Prophylactic oral acyclovir after renal transplantation. Transplantation. 1985;39:279–281. doi: 10.1097/00007890-198503000-00014. [DOI] [PubMed] [Google Scholar]
- 97.Tyring SK, Baker D, Snowden W. Valacyclovir for herpes simples virus infection: long-term safety and sustained efficacy after 20 years’ experience with acyclovir. J Infect Dis. 2002;186(Suppl 1):S40–S46. doi: 10.1086/342966. [DOI] [PubMed] [Google Scholar]
- 98.Levin MJ, Bacon TH, Leary JJ. Resistance of herpes simplex virus infections to nucleoside analogues in HIV-infected patients. Clin Infect Dis. 2004;39(Suppl 5):S248–S257. doi: 10.1086/422364. [DOI] [PubMed] [Google Scholar]
- 99.Safrin S. A controlled trial comparing foscarnet with vidarabine for acyclovir-resistant mucocutaneous herpes simplex infection in the AIDS patient. N Engl J Med. 1991;325:551–555. doi: 10.1056/NEJM199108223250805. [DOI] [PubMed] [Google Scholar]
- 100.Kasper WJ, Howe PM. Fatal varicella after a single course of corticosteroids. Pediatr Infect Dis J. 1990;9:729–732. doi: 10.1097/00006454-199010000-00009. [DOI] [PubMed] [Google Scholar]
- 101.Al-Hoamoudi WK. Severe autoimmune hepatitis triggered by varicella zoster infection. World J Gastroenterol. 2009;15:1004–1006. doi: 10.3748/wjg.15.1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Sample J. Epstein-Barr virus strains differ in their EBNA-3A, EBNA-3B, and EBNA-3C genes. J Virol. 1990;64:4084–4092. doi: 10.1128/jvi.64.9.4084-4092.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Nicholas J. Analysis of nucleotide sequence of the rightmost 43 kbp of herpesvirus saimiri/HVS) L-DNA: general conservation of genetic organization between HVS and Epstein-Barr virus. Virology. 1992;188:296–310. doi: 10.1016/0042-6822(92)90759-i. [DOI] [PubMed] [Google Scholar]
- 104.Goldberg GN. In utero Epstein-Barr virus (infectious mononucleosis) infection. JAMA. 1981;246:1579–1581. [PubMed] [Google Scholar]
- 105.Peter J, Ray CG. Infectious mononucleosis. Pediatr Rev. 1998;19:276–279. doi: 10.1542/pir.19-8-276. [DOI] [PubMed] [Google Scholar]
- 106.Purtilo DT, Sakamoto K. Epstein-Barr virus and human disease: immune response determines the clinical and pathological expression. Hum Pathol. 1981;12:677–679. doi: 10.1016/s0046-8177(81)80162-1. [DOI] [PubMed] [Google Scholar]
- 107.Kilpatrick ZM. Structural and functional abnormalities of liver in infectious mononucleosis. Arch Intern Med. 1966;117:47–53. [PubMed] [Google Scholar]
- 108.Seemayer TA. X-linked lymphoproliferative disease: twenty-five years after discovery. Pediatr Res. 1995;38:471–478. doi: 10.1203/00006450-199510000-00001. [DOI] [PubMed] [Google Scholar]
- 109.Chang MY, Wallace G, Campbell WG. Fatal infectious mononucleosis. Association with liver necrosis and herpes-like virus particles. Arch Pathol Lab Med. 1975;99:185–191. [PubMed] [Google Scholar]
- 110.Wilson WH. Association of lymphomatoid granulomatosis with Epstein-Barr viral infection of B lymphocytes and response to interferon-alpha 2b. Blood. 1996;87:4531–4537. [PubMed] [Google Scholar]
- 111.Beissner RS, Rappaport ES, Diaz JA. Fatal case of Epstein-Barr virus–associated lymphoproliferative disorder associated with a human immunodeficiency virus infection. Arch Pathol Lab Med. 1987;111:250–253. [PubMed] [Google Scholar]
- 112.Albrecht H, Schäfer H, Stellbrinck H-J, Greten H. Epstein-Barr-virus–associated hemophagocytic syndrome. A cause of fever of unknown origin in HIV infection. Arch Pathol Lab Med. 1997;121:853–858. [PubMed] [Google Scholar]
- 113.Hanto DW. Classification of Epstein-Barr virus associated posttransplant lymphoproliferative diseases: implications for understanding their pathogenesis and developing rational treatment strategies. Annu Rev Med. 1995;36:381–394. doi: 10.1146/annurev.med.46.1.381. [DOI] [PubMed] [Google Scholar]
- 114.Capello D. Analysis of immunoglobulin heavy and light chain variable genes in post-transplant lymphoproliferative disorders. Hematol Oncol. 2006;24:212–219. doi: 10.1002/hon.791. [DOI] [PubMed] [Google Scholar]
- 115.Bruu AL. Evaluation of 12 commercial tests for detection of Epstein-Barr virus–specific and heterophile antibodies. Clin Diagn Lab Immunol. 2000;7:451–456. doi: 10.1128/cdli.7.3.451-456.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Weinberger B. Quantitation of Epstein-Barr virus mRNA using reverse transcription and real-time PCR. J Med Virol. 2004;74:612–618. doi: 10.1002/jmv.20220. [DOI] [PubMed] [Google Scholar]
- 117.Torre D, Tambini R. Acyclovir for treatment of infectious mononucleosis: a metaanalysis. Scand J Infect Dis. 1999;31:543–547. doi: 10.1080/00365549950164409. [DOI] [PubMed] [Google Scholar]
- 118.Gross TG. Low dose chemotherapy of Epstein-Barr virus–positive post-transplantation lymphoproliferative disease in children after solid organ transplantation. J Clin Oncol. 2005;23:6481–6488. doi: 10.1200/JCO.2005.08.074. [DOI] [PubMed] [Google Scholar]
- 119.Fohrer C. Long-term survival in post-transplant lymphoproliferative disorders with a dose-adjusted ACVBP regimen. Br J Haematol. 2006;134:602–612. doi: 10.1111/j.1365-2141.2006.06228.x. [DOI] [PubMed] [Google Scholar]
- 120.Verschuuren EA. Treatment of posttransplant lymphoproliferative disease with rituximab: the remission, the relapse, and the complication. Transplantation. 2002;73:100–104. doi: 10.1097/00007890-200201150-00019. [DOI] [PubMed] [Google Scholar]
- 121.Oertel SH. Effect of anti-CD20 antibody rituximab in patients with post-transplant lymphoproliferative disorder (PTLD) Am J Transplant. 2005;5:2901–2906. doi: 10.1111/j.1600-6143.2005.01098.x. [DOI] [PubMed] [Google Scholar]
- 122.Choquet S. Efficacy and safety of rituximab in B-cell post-transplantation lymphoproliferative disorders: results of a prospective multicentre phase 2 study. Blood. 2006;107:3053–3057. doi: 10.1182/blood-2005-01-0377. [DOI] [PubMed] [Google Scholar]
- 123.Rees L, Thomas A, Amlot PL. Disappearance of an Epstein-Barr virus–positive posttransplant plasmacytoma with reduction of immunosuppression. Lancet. 1998;352:789. doi: 10.1016/S0140-6736(05)60684-8. [DOI] [PubMed] [Google Scholar]
- 124.Snover DC, Horwitz CA. Liver disease in cytomegalovirus mononucleosis: a light microscopical and immunoperoxidase study of six cases. Hepatology. 1984;4:408–412. doi: 10.1002/hep.1840040309. [DOI] [PubMed] [Google Scholar]
- 125.Clarke J. Cytomegalovirus granulomatous hepatitis. Am J Med. 1979;66:264–269. doi: 10.1016/0002-9343(79)90543-6. [DOI] [PubMed] [Google Scholar]
- 126.Kylat RI, Kelly EN, Ford-Jones EL. Clinical findings and adverse outcome in neonates with symptomatic congenital cytomegalovirus (SCCMV) infection. Eur J Pediatr. 2006;165:773–778. doi: 10.1007/s00431-006-0172-6. [DOI] [PubMed] [Google Scholar]
- 127.Finegold MJ, Carpenter RJ. Obliterative cholangitis due to cytomegalovirus: a possible precursor of paucity of intrahepatic bile ducts. Hum Pathol. 1982;13:662–665. doi: 10.1016/s0046-8177(82)80011-7. [DOI] [PubMed] [Google Scholar]
- 128.Vanstapel M-J, Desmet VJ. Cytomegalovirus hepatitis: a histological and immunohistochemical study. Appl Pathol. 1983;1:41–49. [PubMed] [Google Scholar]
- 129.Palmer M, Braly LF, Schaffner F. The liver in AIDS disease. Semin Liver Dis. 1987;7:192–202. doi: 10.1055/s-2008-1040576. [DOI] [PubMed] [Google Scholar]
- 130.Dowsett JF. Sclerosing cholangitis in acquired immunodeficiency syndrome. Case reports and a review of the literature. Scand J Gastroenterol. 1988;23:1267–1274. doi: 10.3109/00365528809090203. [DOI] [PubMed] [Google Scholar]
- 131.Cello JP. Acquired immunodeficiency syndrome cholangiopathy: spectrum of disease. Am J Med. 1989;86:539–546. doi: 10.1016/0002-9343(89)90381-1. [DOI] [PubMed] [Google Scholar]
- 132.Bonacini M. Hepatobiliary complications in patients with human immunodeficiency virus infection. Am J Med. 1992;92:404–411. doi: 10.1016/0002-9343(92)90271-c. [DOI] [PubMed] [Google Scholar]
- 133.Shinkai M. Utility of urine and leukocyte cultures and plasma DNA polymerase chain reaction for identification of AIDS patients at risk for developing cytomegalovirus disease. J Infect Dis. 1997;175:302–308. doi: 10.1093/infdis/175.2.302. [DOI] [PubMed] [Google Scholar]
- 134.Humar A. Clinical utility of quantitative cytomegalovirus viral load determination for predicting cytomegalovirus disease in liver transplant recipients. Transplantation. 1999;68:1305–1311. doi: 10.1097/00007890-199911150-00015. [DOI] [PubMed] [Google Scholar]
- 135.Caliendo AM. Comparison of molecular tests for detection and quantification of cell-associated cytomegalovirus DND. J Clin Microbiol. 2003;41:3509–3513. doi: 10.1128/JCM.41.8.3509-3513.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Razonable RR. The clinical use of various blood compartments for cytomegalovirus (CMV) DNA quantitation in transplant recipients with CMV disease. Transplantation. 2002;73:968–973. doi: 10.1097/00007890-200203270-00025. [DOI] [PubMed] [Google Scholar]
- 137.Goodrich JM, Boeckh M, Bowden R. Strategies for the prevention of cytomegalovirus disease after bone marrow transplantation. Clin Infect Dis. 1994;19:287–298. doi: 10.1093/clinids/19.2.287. [DOI] [PubMed] [Google Scholar]
- 138.Paya C. Prevention of cytomegalovirus disease in recipients of solid-organ transplants. Clin Infect Dis. 2001;32:596–603. doi: 10.1086/318724. [DOI] [PubMed] [Google Scholar]
- 139.Crumpacker CS. Ganciclovir. N Engl J Med. 1996;335:721–729. doi: 10.1056/NEJM199609053351007. [DOI] [PubMed] [Google Scholar]
- 140.Martin BK. Change over time in incidence of ganciclovir resistance in patients with cytomegalovirus retinitis. Clin Infect Dis. 2008;44:1001–1008. doi: 10.1086/512368. [DOI] [PubMed] [Google Scholar]
- 141.Santoro F. CD46 is a cellular receptor for human herpesvirus 6. Cell. 1999;99:817–827. doi: 10.1016/s0092-8674(00)81678-5. [DOI] [PubMed] [Google Scholar]
- 142.Leach CT. Human herpesvirus-6 and -7 infections in children: agents of roseola and other syndromes. Curr Opin Pediatr. 2000;12:269–274. doi: 10.1097/00008480-200006000-00017. [DOI] [PubMed] [Google Scholar]
- 143.Stoeckle MY. The spectrum of human herpesvirus 6 infection: from roseola infantum to adult disease. Annu Rev Med. 2000;51:423–430. doi: 10.1146/annurev.med.51.1.423. [DOI] [PubMed] [Google Scholar]
- 144.Asano Y. Fatal fulminant hepatitis in an infant with herpesvirus-6 infection. Lancet. 1990;335:862–863. doi: 10.1016/0140-6736(90)90983-c. [DOI] [PubMed] [Google Scholar]
- 145.Mendel I. Fulminant hepatitis in neonates with human herpesvirus 6 infection. Pediatr Infect Dis J. 1995;14:993–997. doi: 10.1097/00006454-199511000-00013. [DOI] [PubMed] [Google Scholar]
- 146.Chevret L. Human herpesvirus-6 infection: a prospective study evaluating HHV-6 DNA levels in liver from children with acute liver failure. J Med Virol. 2008;80:1051–1057. doi: 10.1002/jmv.21143. [DOI] [PubMed] [Google Scholar]
- 147.Härmä M, Höckerstedt K, Lautenschlager I. Human herpesvirus-6 and acute liver failure. Transplantation. 2003;76:536–539. doi: 10.1097/01.TP.0000069233.13409.DF. [DOI] [PubMed] [Google Scholar]
- 148.Nobili V. Acute liver failure as presenting feature in tyrosinemia type 1 in a child with primary HHV-6 infection. J Gastroenterol Hepatol. 2006;21:339. doi: 10.1111/j.1440-1746.2006.04045.x. [DOI] [PubMed] [Google Scholar]
- 149.Chang YL. Human herpesvirus 6–related fulminant myocarditis in an immunocompetent adult with fatal outcome. Hum Pathol. 2009;40:740–745. doi: 10.1016/j.humpath.2008.08.017. [DOI] [PubMed] [Google Scholar]
- 150.Cacheux W. HHV-6–related acute liver failure in two immunocompetent adults: favourable outcome after liver transplantation and/or ganciclovir therapy. J Intern Med. 2005;258:573–578. doi: 10.1111/j.1365-2796.2005.01567.x. [DOI] [PubMed] [Google Scholar]
- 151.Yoshikawa T. A prospective study of human herpesvirus-6 infection in renal transplantation. Transplantation. 1992;54:879–883. doi: 10.1097/00007890-199211000-00022. [DOI] [PubMed] [Google Scholar]
- 152.Ward KN, Gray JJ, Efstathiou S. Brief report: primary human herpes virus 6 infection in a patient following liver transplantation from a seropositive donor. J Med Virol. 1989;28:69–72. doi: 10.1002/jmv.1890280203. [DOI] [PubMed] [Google Scholar]
- 153.Dockrell DH, Paya CV. Human herpesvirus-6 and -7 in transplantation. Rev Med Virol. 2001;11:23–36. doi: 10.1002/rmv.299. [DOI] [PubMed] [Google Scholar]
- 154.Härmä M. Pre-transplant human herpesvirus 6 infection of patients with acute liver failure is a risk factor for post transplant human herpesvirus 6 infection of the liver. Transplantation. 2006;81:367–372. doi: 10.1097/01.tp.0000195771.83614.0b. [DOI] [PubMed] [Google Scholar]
- 155.Ohashi M. Human herpesvirus 6 infection in adult living related liver transplant recipients. Liver Transpl. 2008;14:100–109. doi: 10.1002/lt.21304. [DOI] [PubMed] [Google Scholar]
- 156.Tanaka-Taya K. Reactivation of human herpesvirus 6 by infection of human herpesvirus 7. J Med Virol. 2000;60:284–289. doi: 10.1002/(sici)1096-9071(200003)60:3<284::aid-jmv6>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
- 157.Ihira M. Correlation between human herpesvirus 6 and 7 infections after living related liver transplantation. Microbiol Immunol. 2001;45:225–232. doi: 10.1111/j.1348-0421.2001.tb02611.x. [DOI] [PubMed] [Google Scholar]
- 158.Humar A. Human herpesvirus-6 is associated with cytomegalovirus reactivation in liver transplant recipients. J Infect Dis. 2000;181:1450–1453. doi: 10.1086/315391. [DOI] [PubMed] [Google Scholar]
- 159.Potenza I, Luppi M, Barozzi P. HHV-6A in syncytial giant-cell hepatitis. N Engl J Med. 2008;359:583–602. doi: 10.1056/NEJMoa074479. [DOI] [PubMed] [Google Scholar]
- 160.Kuntzen T. Postinfantile giant cell hepatitis with autoimmune features following a human herpesvirus 6–induced adverse drug reaction. Eur J Gastroenterol Hepatol. 2005;17:1131–1134. doi: 10.1097/00042737-200510000-00020. [DOI] [PubMed] [Google Scholar]
- 161.Randhawa PS. Herpesvirus 6 variant A infection after heart transplantation with giant cell transformation in bile ductular and gastroduodenal epithelium. Am J Surg Pathol. 1997;21:847–853. doi: 10.1097/00000478-199707000-00014. [DOI] [PubMed] [Google Scholar]
- 162.Suga S. IgM neutralizing antibody responses to human herpesvirus-6 in patients with exanthema subitum or organ transplantation. Microbiol Immunol. 1992;36:495–506. doi: 10.1111/j.1348-0421.1992.tb02047.x. [DOI] [PubMed] [Google Scholar]
- 163.Deback C. Detection of human herpesviruses HHV-6, HHV-7 and HHV-8 in whole blood by real-time PCR using the new CMV, HHV-6, 7, 8 R-gene kit. J Virol Methods. 2008;149:285–291. doi: 10.1016/j.jviromet.2008.01.026. [DOI] [PubMed] [Google Scholar]
- 164.Engelmann I. Rapid quantitative PCR assay for the simultaneous detection of herpes simplex virus, varicella zoster virus, cytomegalovirus, Epstein-Barr virus, and human herpesvirus 6 DNA in blood and other clinical specimens. J Med Virol. 2008;80:467–477. doi: 10.1002/jmv.21095. [DOI] [PubMed] [Google Scholar]
- 165.Flamand L. Multicenter comparison of PCR assays for detection of human herpesvirus 6 DNA in serum. J Clin Microbiol. 2008;46:2700–2706. doi: 10.1128/JCM.00370-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Yoshida M. Comparison of antiviral compounds against human herpesvirus 6 and 7. Antiviral Res. 1998;40:73–84. doi: 10.1016/s0166-3542(98)00049-7. [DOI] [PubMed] [Google Scholar]
- 167.Isegawa Y. Human herpesvirus 6 ganciclovir-resistant strain with amino acid substitutions associated with the death of an allogeneic stem cell transplant recipient. J Clin Virol. 2009;44:15–19. doi: 10.1016/j.jcv.2008.09.002. [DOI] [PubMed] [Google Scholar]
- 168.De Clercq E. Antiviral agents against human herpesviruses HHV-6, HHV-7 and HHV-8. Rev Med Virol. 2001;11:381–395. doi: 10.1002/rmv.336. [DOI] [PubMed] [Google Scholar]
- 169.Tokimasa S. Ganciclovir is effective for prophylaxis and treatment of human herpesvirus-6 in allogeneic stem cell transplantation. Bone Marrow Transplant. 2002;29:595–598. doi: 10.1038/sj.bmt.1703423. [DOI] [PubMed] [Google Scholar]
- 170.Bonnafous P. Characterization of a cidofovir-resistant HHV-6 mutant obtained by in vitro selection. Antiviral Res. 2008;77:237–240. doi: 10.1016/j.antiviral.2007.12.004. [DOI] [PubMed] [Google Scholar]
- 171.Lee LM, Henderson DK. Emerging viral infections. Curr Opin Infect Dis. 2001;14:467–480. doi: 10.1097/00001432-200108000-00012. [DOI] [PubMed] [Google Scholar]
- 172.Wijnveen AC. Disseminated Kaposi's sarcoma—full regression after withdrawal of immunosuppressive therapy: report of a case. Transplant Proc. 1987;19:3735–3736. [PubMed] [Google Scholar]
- 173.Nagy S. Iatrogenic Kaposi's sarcoma: HHV8 positivity persists but tumors regress almost completely without immunosuppressive therapy. Transplantation. 2000;69:2230–2231. doi: 10.1097/00007890-200005270-00053. [DOI] [PubMed] [Google Scholar]
- 174.Paparizos VA. Response of AIDS-associated Kaposi sarcoma to highly active antiretroviral therapy alone. J Acquir Immune Def Syndr. 2002;30:257–258. doi: 10.1097/00042560-200206010-00015. [DOI] [PubMed] [Google Scholar]
- 175.Engles EA. Risk factor for human herpesvirus 8 infection among adults in the United States and evidence for sexual transmission. J Infect Dis. 2007;196:199–207. doi: 10.1086/518791. [DOI] [PubMed] [Google Scholar]
- 176.Piselli P. Risk of Kaposi sarcoma after solid-organ transplantation: multicenter study in 4,767 recipients in Italy, 1970-2006. Transplant Proc. 2009;41:1227–1230. doi: 10.1016/j.transproceed.2009.03.009. [DOI] [PubMed] [Google Scholar]
- 177.Hasan FA. Hepatic involvement as the primary manifestation of Kaposi sarcoma in the acquired immune deficiency syndrome. Am J Gastroenterol. 1989;84:1449–1451. [PubMed] [Google Scholar]
- 178.Glasgow BJ. Clinical and pathologic findings of the liver in acquired immune deficiency syndrome (AIDS) Am J Clin Pathol. 1985;83:482–488. doi: 10.1093/ajcp/83.5.582. [DOI] [PubMed] [Google Scholar]
- 179.Naresh KN, Rice AJ, Bower M. Lymph nodes involved by multicentric Castleman disease among HIV-positive individuals are often involved by Kaposi sarcoma. Am J Surg Pathol. 2008;32:1006–1012. doi: 10.1097/PAS.0b013e318160ed97. [DOI] [PubMed] [Google Scholar]
- 180.Koreishi A. Synchronous follicular lymphoma, Kaposi sarcoma, and Castleman's disease in a HIV-negative patient with EBV and HHV-8 coinfection. Int J Surg Pathol. 2009 doi: 10.1177/1066896909341803. Aug 5 [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- 181.Cesarman E, Knowles DM. The role of Kaposi's sarcoma–associated herpesvirus (HSHV/HHV8) in lymphoproliferative diseases. Semin Cancer Biol. 1999;9:165–174. doi: 10.1006/scbi.1998.0118. [DOI] [PubMed] [Google Scholar]
- 182.Campistol JM, Schena FP. Kaposi's sarcoma in renal transplant recipients—the impact of proliferation signal inhibitors. Nephrol Dial Transplant. 2007;22(Suppl 1):17–22. doi: 10.1093/ndt/gfm089. [DOI] [PubMed] [Google Scholar]
- 183.Barozzi P. Indirect antitumor effects of mammalian target of rapamycin inhibitors against Kaposi sarcoma in transplant patients. Transplantation. 2009;88:597–598. doi: 10.1097/TP.0b013e3181b15d56. [DOI] [PubMed] [Google Scholar]
- 184.Babel N. Development of Kaposi's sarcoma under sirolimus-based immunosuppression and successful treatment with imiquimod. Transpl Infect Dis. 2008;10:59–62. doi: 10.1111/j.1399-3062.2007.00239.x. [DOI] [PubMed] [Google Scholar]
- 185.Van der Ende M. Complete clinical and virological remission of refractory HIV-related Kaposi's sarcoma with pegylated interferon alpha. AIDS. 2007;21:1661–1662. doi: 10.1097/QAD.0b013e32827035a4. [DOI] [PubMed] [Google Scholar]
- 186.Di Trolio R. Role of pegylated liposomal doxorubicin (PLD) in systemic Kaposi's sarcoma: a systematic review. Int J Immunopathol Pharmacol. 2006;19:253–263. doi: 10.1177/039463200601900202. [DOI] [PubMed] [Google Scholar]
- 187.Stebbing J. Paclitaxel for anthracycline-resistant AIDS-related Kaposi's sarcoma: clinical and angiogenic correlations. Ann Oncol. 2003;14:1660–1666. doi: 10.1093/annonc/mdg461. [DOI] [PubMed] [Google Scholar]
- 188.Bower M. Brief communication: rituximab in HIV-associated multicentric Castleman disease. Ann Intern Med. 2007;147:836–839. doi: 10.7326/0003-4819-147-12-200712180-00003. [DOI] [PubMed] [Google Scholar]
- 189.Casper C. Valganciclovir for suppression of human herpesvirus-8 replication: a randomized, double-blind, placebo-controlled, crossover trial. J Infect Dis. 2008;198:23–30. doi: 10.1086/588820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Garnett CT. Prevalence and quantitation of species C adenovirus DNA in human mucosal lymphocytes. J Virol. 2002;76:10608–10616. doi: 10.1128/JVI.76.21.10608-10616.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Garnett CT. Latent species C adenoviruses in human tonsil tissues. J Virol. 2009;83:2417–2428. doi: 10.1128/JVI.02392-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Kojaoghlanian T, Flomenberg P, Horwitz MS. The impact of adenovirus infection on the immunocompromised host. Rev Med Virol. 2003;13:155–171. doi: 10.1002/rmv.386. [DOI] [PubMed] [Google Scholar]
- 193.Carmichael GP. Adenovirus hepatitis in an immunosuppressed adult patient. Am J Clin Pathol. 1979;71:352–355. doi: 10.1093/ajcp/71.3.352. [DOI] [PubMed] [Google Scholar]
- 194.South MA. Fatal adenovirus hepatic necrosis in severe combined immune deficiency. Pediatr Infect Dis. 1982;1:416–419. doi: 10.1097/00006454-198211000-00012. [DOI] [PubMed] [Google Scholar]
- 195.Janner D. Fatal adenovirus infection in a child with acquired immunodeficiency syndrome. Pediatr Infect Dis. 1990;9:434–436. doi: 10.1097/00006454-199006000-00013. [DOI] [PubMed] [Google Scholar]
- 196.Krilov LR. Disseminated adenovirus infection with hepatic necrosis in patients with human immunodeficiency virus infection and other immunodeficiency states. Rev Infect Dis. 1990;12:303–307. doi: 10.1093/clinids/12.2.303. [DOI] [PubMed] [Google Scholar]
- 197.Michaels MG. Adenovirus infection in pediatric liver transplant recipients. J Infect Dis. 1992;165:170–174. doi: 10.1093/infdis/165.1.170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Koneru B. Serological studies of adenoviral hepatitis following pediatric liver transplantation. Transplant Proc. 1990;22:1547–1548. [PMC free article] [PubMed] [Google Scholar]
- 199.Wulffraat NM. Recovery from adenovirus pneumonia in a severe combined immunodeficiency patient with intravenous ribavirin. Transplantation. 1995;59:927. [PubMed] [Google Scholar]
- 200.Polakos NK. Kupffer cell–dependent hepatitis occurs during influenza infection. Am J Pathol. 2006;168:1169–1178. doi: 10.2353/ajpath.2006.050875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Schumann J. Importance of Kupffer cells for T-cell–dependent liver injury in mice. Am J Pathol. 2000;157:1671–1683. doi: 10.1016/S0002-9440(10)64804-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Murali-Krishna K. Counting antigen-specific CD8 T cells: a reevaluation of bystander activation during viral infection. Immunity. 1998;8:177–187. doi: 10.1016/s1074-7613(00)80470-7. [DOI] [PubMed] [Google Scholar]
- 203.Yuen KY, Wong SSY. Human infection by avian influenza A H5N1. Hong Kong Med J. 2005;11:189–199. [PubMed] [Google Scholar]
- 204.To KF. Pathology of fatal human infection associated with avian influenza A H5N1 virus. J Med Virol. 2001;63:242–246. doi: 10.1002/1096-9071(200103)63:3<242::aid-jmv1007>3.0.co;2-n. [DOI] [PubMed] [Google Scholar]
- 205.Peiris JS. Re-emergence of fatal human influenza A subtype H5N1 disease. Lancet. 2004;363:617–619. doi: 10.1016/S0140-6736(04)15595-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Eisenhut M, Thorburn K, Ahmed T. Transaminase levels in ventilated children with respiratory syncytial virus bronchiolitis. Intensive Care Med. 2004;30:931–934. doi: 10.1007/s00134-004-2236-2. [DOI] [PubMed] [Google Scholar]
- 207.Eisenhut M. Ischemic hepatitis and collateral damage to the liver in severe viral respiratory tract infections. Am J Pathol. 2006;169:1100. doi: 10.2353/ajpath.2006.060514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Ksiazek TG. A novel coronavirus in patients with severe acute respiratory syndrome. N Engl J Med. 2003;348:1953–1966. doi: 10.1056/NEJMoa030781. [DOI] [PubMed] [Google Scholar]
- 209.Drosten C. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N Engl J Med. 2003;348:1967–1976. doi: 10.1056/NEJMoa030747. [DOI] [PubMed] [Google Scholar]
- 210.Lee N. A major outbreak of severe acute respiratory syndrome in Hong Kong. N Engl J Med. 2003;348:1986–1994. doi: 10.1056/NEJMoa030685. [DOI] [PubMed] [Google Scholar]
- 211.Booth CM. Clinical features and short term outcome of 144 patients with SARS in the greater Toronto area. JAMA. 2003;289:2801–2809. doi: 10.1001/jama.289.21.JOC30885. [DOI] [PubMed] [Google Scholar]
- 212.Poutanen SM. Identification of severe acute respiratory syndrome in Canada. N Engl J Med. 2003;348:1995–2005. doi: 10.1056/NEJMoa030634. [DOI] [PubMed] [Google Scholar]
- 213.Choi KW. Outcomes and prognostic factors in 267 patients with severe acute respiratory syndrome in Hong Kong. Ann Intern Med. 2003;139:715–723. doi: 10.7326/0003-4819-139-9-200311040-00005. [DOI] [PubMed] [Google Scholar]
- 214.Wong WM. Temporal pattern of hepatic dysfunction and disease severity in patients with SARS. JAMA. 2003;290:2663–2665. doi: 10.1001/jama.290.20.2663. [DOI] [PubMed] [Google Scholar]
- 215.Chan HL-Y. Clinical significance of hepatic derangement in severe acute respiratory syndrome. World J Gastroenterol. 2005;11:2148–2153. doi: 10.3748/wjg.v11.i14.2148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Chau TN. SARS-associated viral hepatitis caused by a novel coronavirus: report of three cases. Hepatology. 2004;39:302–310. doi: 10.1002/hep.20111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Shi X. Severe acute respiratory syndrome–associated coronavirus is detected in intestinal tissues of fatal cases. Am J Gastroenterol. 2005;100:169–176. doi: 10.1111/j.1572-0241.2005.40377.x. [DOI] [PubMed] [Google Scholar]
- 218.Hsiao CH. Immunohistochemical study of severe acute respiratory syndrome–associated coronavirus in tissue sections of patients. J Formos Med Assoc. 2005;104:150–156. [PubMed] [Google Scholar]
- 219.Lo AW, Tang NLS, To KF. How the SARS coronavirus causes disease: host or organism? J Pathol. 2006;208:142–151. doi: 10.1002/path.1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Kaplan LJ. Severe measles in immunocompromised patients. JAMA. 1992;267:1237–1241. [PubMed] [Google Scholar]
- 221.Siegel D, Hirschman SZ. Hepatic dysfunction in acute measles infection of adults. Arch Intern Med. 1977;137:1178–1179. [PubMed] [Google Scholar]
- 222.Nickell MD, Cannady PB, Schwitzer GA. Subclinical hepatitis in rubeola infections in young adults. Ann Intern Med. 1979;90:354–355. doi: 10.7326/0003-4819-90-3-354. [DOI] [PubMed] [Google Scholar]
- 223.Gavish D, Kleinman Y, Morag A, Chajek-Shaul T. Hepatitis and jaundice associated with measles in adults. Arch Intern Med. 1983;143:674–677. [PubMed] [Google Scholar]
- 224.Frey HM. Atypical measles syndrome: unusual hepatic, pulmonary, and immunologic aspects. Am J Med Sci. 1981;281:51–55. doi: 10.1097/00000441-198101000-00008. [DOI] [PubMed] [Google Scholar]
- 225.McLellan RK, Gleiner JA. Acute hepatitis in an adult with rubeola. JAMA. 1982;247:2000–2001. [PubMed] [Google Scholar]
- 226.Shalev-Zimels H. Extent of measles hepatitis in various ages. Hepatology. 1988;8:1138–1139. doi: 10.1002/hep.1840080529. [DOI] [PubMed] [Google Scholar]
- 227.Khatib R, Siddique M, Abbass M. Measles associated hepatobiliary disease: an overview. Infection. 1993;21:112–114. doi: 10.1007/BF01710744. [DOI] [PubMed] [Google Scholar]
- 228.Satoh A. Clinicopathological study on liver dysfunction in measles. Intern Med. 1999;38:454–457. doi: 10.2169/internalmedicine.38.454. [DOI] [PubMed] [Google Scholar]
- 229.Scully RE, Mark EJ, McNeely WF, McNeely BU. Case records of the Massachusetts General Hospital [measles hepatitis] N Engl J Med. 1988;319:495–509. [Google Scholar]
- 230.Robertson DAF, Zhang SL, Guy EC, Wright R. Persistent measles virus genome in autoimmune chronic active hepatitis. Lancet. 1987;ii:9–11. doi: 10.1016/s0140-6736(87)93051-0. [DOI] [PubMed] [Google Scholar]
- 231.Phillips MJ. Syncytial giant-cell hepatitis: sporadic hepatitis with distinctive pathological features, a severe clinical course and paramyxoviral features. N Engl J Med. 1991;324:455–460. doi: 10.1056/NEJM199102143240705. [DOI] [PubMed] [Google Scholar]
- 232.Fimmerl CJ. A case of syncytial giant cell hepatitis with features of a paramyxoviral infection. Am J Gastroenterol. 1998;93:1931–1937. doi: 10.1111/j.1572-0241.1998.00548.x. [DOI] [PubMed] [Google Scholar]
- 233.Hicks J, Barrish J, Zhu SH. Neonatal syncytial giant cell hepatitis with paramyxoviral-like inclusions. Ultrastruct Pathol. 2001;25:65–71. doi: 10.1080/019131201300004708. [DOI] [PubMed] [Google Scholar]
- 234.Alexopoulou A. A fatal case of postinfantile giant cell hepatitis in a patient with chronic lymphocytic leukaemia. Eur J Gastroenterol Hepatol. 2003;15:551–555. doi: 10.1097/01.meg.0000050026.34359.7c. [DOI] [PubMed] [Google Scholar]
- 235.Stern H, Williams BM. Isolation of rubella virus in a case of neonatal giant-cell hepatitis. Lancet. 1966;i:293–295. doi: 10.1016/s0140-6736(66)90642-8. [DOI] [PubMed] [Google Scholar]
- 236.Hosier DM, Newton WA. Serious Coxsackie infection in infants and children: myocarditis, meningoencephalitis, and hepatitis. J Dis Child. 1958;96:251–267. doi: 10.1001/archpedi.1958.02060060253001. [DOI] [PubMed] [Google Scholar]
- 237.O'Shaughnessy WJ, Buechner HA. Hepatitis associated with Coxsackie B5 virus infection during late pregnancy. JAMA. 1962;179:71–72. doi: 10.1001/jama.1962.03050010000017. [DOI] [PubMed] [Google Scholar]
- 238.Sun NC, Smith VM. Hepatitis associated with myocarditis. Unusual manifestation of infection with Coxsackie virus group B, type 3. N Engl J Med. 1966;274:190–193. doi: 10.1056/NEJM196601272740405. [DOI] [PubMed] [Google Scholar]
- 239.Hayakawa H. A clinical study of adult human parvovirus B 19 infection. Intern Med. 2007;41:295–299. doi: 10.2169/internalmedicine.41.295. [DOI] [PubMed] [Google Scholar]
- 240.Waza K, Inoue K, Matsumura S. Symptoms associated with parvovirus B19 infection in adults: a pilot study. Intern Med. 2007;46:1975–1978. doi: 10.2169/internalmedicine.46.0366. [DOI] [PubMed] [Google Scholar]
- 241.Anand A. Human parvovirus infection in pregnancy and hydrops fetalis. N Engl J Med. 1987;316:183–186. doi: 10.1056/NEJM198701223160403. [DOI] [PubMed] [Google Scholar]
- 242.Yoto Y. Human parvovirus B19 infection associated with acute hepatitis. Lancet. 1996;347:868–869. doi: 10.1016/s0140-6736(96)91348-3. [DOI] [PubMed] [Google Scholar]
- 243.Langnas AN. Parvovirus B19 as a possible causative agent of fulminant liver failure and associated aplastic anemia. Hepatology. 1995;22:1661–1665. [PubMed] [Google Scholar]
- 244.Lee WM. Brief report: no evidence for parvovirus B19 or hepatitis E as a cause of acute liver failure. Dig Dis Sci. 2006;51:1712–1715. doi: 10.1007/s10620-005-9061-5. [DOI] [PubMed] [Google Scholar]
- 245.Nishizawa T. A novel DNA virus (TTV) associated with elevated transaminases in posttransfusion hepatitis of unknown etiology. Biochem Biophys Res Commun. 1997;241:92–99. doi: 10.1006/bbrc.1997.7765. [DOI] [PubMed] [Google Scholar]
- 246.Mushahwar IK. Molecular and biophysical characterization of TT virus: evidence for a new virus family infecting humans. Proc Natl Acad Sci U S A. 1999;96:3177–3182. doi: 10.1073/pnas.96.6.3177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Kanda Y. TT virus in bone marrow transplant recipients. Blood. 1999;93:2485–2490. [PubMed] [Google Scholar]
- 248.Matsumoto A. Transfusion-associated TT virus infection and its relationship to liver disease. Hepatology. 1999;30:283–288. doi: 10.1002/hep.510300118. [DOI] [PubMed] [Google Scholar]
- 249.Parquet MC. Prevalence and clinical characteristics of TT virus (TTV) in patients with sporadic acute hepatitis of unknown etiology. J Hepatol. 1999;31:985–989. doi: 10.1016/s0168-8278(99)80309-2. [DOI] [PubMed] [Google Scholar]
- 250.Piaggio F. Torqueteno virus—cause of viral liver disease following liver transplantation: a case report. Transplant Proc. 2009;41:1378–1379. doi: 10.1016/j.transproceed.2009.03.047. [DOI] [PubMed] [Google Scholar]
- 251.Foschini MF. TT virus–related acute recurrent hepatitis. Histological features of a case and review of the literature. Virchows Arch. 2001;439:752–755. doi: 10.1007/s004280100467. [DOI] [PubMed] [Google Scholar]
- 252.Takayama S. Persistence of a novel TT virus (TTV) infection in Japanese haemophiliacs. Br J Haematol. 1999;104:626–629. doi: 10.1046/j.1365-2141.1999.01207.x. [DOI] [PubMed] [Google Scholar]
- 253.Naoumov NV. Presence of a newly described human DNA virus (TTV) in patients with liver disease. Lancet. 1998;352:195–197. doi: 10.1016/S0140-6736(98)04069-0. [DOI] [PubMed] [Google Scholar]
- 254.Cleavinger PJ. Prevalence of TT virus infection in blood donors with elevated ALT in the absence of known hepatitis markers. Am J Gastroenterol. 2000;95:772–776. doi: 10.1111/j.1572-0241.2000.01820.x. [DOI] [PubMed] [Google Scholar]
- 255.Charlton M. TTV-virus infection in North American blood donors, patients with fulminant hepatic failure, and cryptogenic cirrhosis. Hepatology. 1998;28:839–842. doi: 10.1002/hep.510280335. [DOI] [PubMed] [Google Scholar]
- 256.Tagger A. A case-control study on a novel DNA virus (TT virus) infection and hepatocellular carcinoma. Hepatology. 1999;30:294–299. doi: 10.1002/hep.510300115. [DOI] [PubMed] [Google Scholar]
- 257.Ikeda H. Infection with an unenveloped DNA virus (TTV) in patients with acute or chronic liver disease of unknown etiology and in those positive for hepatitis C virus RNA. J Hepatol. 1999;30:205–212. doi: 10.1016/s0168-8278(99)80063-4. [DOI] [PubMed] [Google Scholar]
- 258.Nishiguchi S. GB virus C and TT virus infections in Japanese patients with autoimmune hepatitis. J Med Virol. 2002;66:258–262. doi: 10.1002/jmv.2139. [DOI] [PubMed] [Google Scholar]