ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Human herpesvirus-8 infection

Human herpesvirus-8 infection
Literature review current through: Jan 2024.
This topic last updated: Dec 11, 2023.

INTRODUCTION — Human herpesvirus-8 (HHV-8), also known as Kaposi sarcoma-associated herpesvirus (KSHV), is a large deoxyribonucleic acid (DNA) virus belonging to the gamma herpesvirus family. It is endemic worldwide and can cause certain cancers in a minority of individuals with infection.

The virology, pathophysiology, epidemiology, clinical manifestations, diagnosis, and the role for direct anti-viral therapy of HHV-8 infection will be reviewed here. Specific clinical syndromes that are associated with HHV-8, including acquired immunodeficiency syndrome (AIDS)-associated Kaposi sarcoma, Classic Kaposi sarcoma, organ transplant-associated Kaposi sarcoma, HHV-8-associated multicentric Castleman disease (MCD), and primary effusion lymphoma, are discussed elsewhere. (See "AIDS-related Kaposi sarcoma: Clinical manifestations and diagnosis" and "AIDS-related Kaposi sarcoma: Staging and treatment" and "Classic Kaposi sarcoma: Epidemiology, risk factors, pathology, and molecular pathogenesis" and "Classic Kaposi sarcoma: Clinical features, staging, diagnosis, and treatment" and "Malignancy after solid organ transplantation", section on 'Kaposi sarcoma' and "HHV-8/KSHV-associated multicentric Castleman disease" and "Primary effusion lymphoma".)

VIROLOGY — Human herpesvirus-8 (HHV-8) is a large (165 kB) double-stranded DNA virus which, like its closest known relative Epstein-Barr virus (EBV), is a member of the gamma herpesvirus group [1].

HHV-8 is classified as a group 1 biological carcinogenic agent, defined as having sufficient evidence to definitively establish a link to carcinogenicity in humans, by the International Agency for Research on Cancer (IARC) [2,3].

HHV-8 is now established as the cause of several human tumors arising from distinct cells types, including Kaposi sarcoma (KS), primary effusion lymphoma, and, in the setting of human immunodeficiency virus (HIV), almost all cases of multicentric Castleman disease (MCD) [4,5]. Patients can manifest more than one of these tumors. It has also been implicated in an inflammatory syndrome distinct from HHV-8-associated MCD, the Kaposi sarcoma-associated herpesvirus (KSHV)/HHV-8 inflammatory cytokine syndrome (KICS) [6,7].

While HHV-8 associated tumors are most common in individuals with HIV, they have also been reported in other immunosuppressed and older individuals, and occasionally in patients with congenital immunodeficiencies [8,9]. Additionally, hemophagocytic syndrome has been reported after HHV-8 infection in infants with heterozygous mutations in perforin alleles [10].

PATHOGENESIS — Human herpesvirus-8 (HHV-8) can infect a variety of cells, including those of endothelial lineage, monocytes, and B cells. Thus, HHV-8-associated cancers derive from different cells of origin among the range of cells infected by HHV-8.

Like all herpesviruses, HHV-8 infection exhibits both latent and lytic phases, distinguished by their viral gene expression patterns [11]. During the latent phase of infection, only a few viral genes are expressed, while during the lytic phase, the full viral genetic program involving multiple genes is expressed and viral replication occurs along with destruction of the host cell.

Latent phase – In its latent phase, HHV-8 is maintained as a circular episome in the nucleus, which is tethered to the host chromosome, thus maintaining its replication during host cell division. The host cell survives latent infection, which is characterized by extremely limited gene expression [12,13]. The genes expressed in latency are directed to enhance cell survival signals, promote angiogenesis, and redirect the host immune response away from the infected cell [11,13-15]. HHV-8 latent transcripts, including genes and miRNAs, function to subvert host signaling pathways and favor viral persistence. Although the primary function of latency-expressed genes in the normal HHV-8 lifecycle is to enable chronically infected cells to remain undetected in the body, these same genes can also promote tumorigenesis in certain circumstances. In the absence of an effective immune response, HHV-8 manipulates the host cell pathways to thwart cellular defenses and promote tumorigenesis.

Lytic phase – Activation of the virally encoded "lytic switch" gene, RTA, causes HHV-8 to enter its lytic phase, in which the full complement of viral-encoded genes is expressed and the host cellular machinery is redirected to the manufacture and assembly of progeny virions [13]. The host cell is destroyed in this process. HHV-8 lytic genes have effects on the host cell, resulting in reprogramming of cellular metabolism, upregulation of survival pathways, stimulation of angiogenesis and inflammation, and escape from immune control.

Intermittent cycles of viral lytic activation are thought to serve two functions. First, it is during lytic phase that onward infection of additional hosts occurs (see 'Transmission' below). Second, by enabling infection of new cells within the host, these cycles are also thought to be important in maintaining viral persistence in the host. The triggers of lytic activation in vivo remain unclear, whereas in vitro there is evidence that activation of cellular response pathways (eg, hypoxia, oxidative stress, coinfection with HIV) trigger lytic activation [16-19].

There is evidence that some HHV-8 lytic genes also play key roles in tumorigenesis, particularly in HHV-8-associated multicentric Castleman disease (MCD).

HHV-8 encodes more than 85 genes, many of which have the potential to be antigenic. However, antibody responses to viral antigens are variable; certain antigens such as latency associated nuclear antigen (LANA) and capsid antigen K8.1 most reliably elicit strong immune responses. Intensity of immune response may also depend on immunocompetence, as well as the coexistence of an HHV-8-associated malignancy [20].

HHV-8-associated malignancies can be seen as an inadvertent byproduct of HHV-8's survival strategies, particularly those aimed at developing persistent infection and thwarting cellular defenses against viral infection. In the absence of an effective immune response to infection, the risk of HHV-8-associated tumors is substantially increased.

EPIDEMIOLOGY — HHV-8-associated tumors are estimated to account for just under 1 percent of all cancers occurring worldwide [21].

Although human herpesvirus-8 (HHV-8) infection is necessary to cause the associated tumors, the tumors require other cofactors (eg, untreated HIV infection, immunosuppression) in order to develop. The risk of developing HHV-8-associated diseases among individuals with HHV-8 without any contributing cofactors is low: only about 1 in 400 to 1 in 1500 individuals with HHV-8 develop Kaposi sarcoma (KS), with the incidence increasing with age (likely due to waning cellular immunity) [22-24]. In contrast, as many as one half of patients with HIV and HHV-8 coinfection will develop KS in the absence of effective antiretroviral treatment [25,26]. In the United States, up to 80 percent of cases of KS occur in people with HIV, whereas in sub-Saharan Africa, this proportion is thought to be lower [27].

Serologic surveys show that, unlike most other human herpesviruses, HHV-8 is not ubiquitous. Significant variations in prevalence occur geographically and across different behavioral groups.

Geographic distribution – HHV-8 infection is very common in sub-Saharan Africa with seropositivity rates of over 50 percent; moderately prevalent in Mediterranean countries (20 to 30 percent); and much less common (well under 10 percent) in most of Europe, Asia, and the United States [22,28,29]. Even within a country, substantial regional variation can occur. For example, among blood donors in Italy, HHV-8 prevalence was estimated to be 7.3 percent in Northern and Central Italy and 24.6 percent in Southern Italy [30]. High HHV-8 antibody prevalence has also been reported in several smaller distinct populations: for example, 65 percent seroprevalence among Brazilian Native American adults [31]; 33 percent among Papua New Guinean adults [32]; 25 percent among Indigenous Australian adults in some regions of central Australia [32]. The reasons for this geographic variation remain unclear. Viral subtype variation across regions is limited but could be playing a role, while both environmental and host genetic factors have been explored without clear roles yet being established for either [33].

Other populations with increased prevalence – HHV-8 prevalence also is elevated in certain behavioral groups regardless of geography [34]. Most strikingly, its prevalence is greatly elevated in men who have sex with men (MSM), independent of the increase in HIV prevalence also seen in that group [28]. Compared with a prevalence of approximately 5 percent in the general United States population, in American MSM, prevalence estimates range from 25 to 60 percent among MSM with HIV and 20 to 30 percent among MSM without HIV [35-38]. HHV-8 infection in MSM without HIV in other countries also appears common [39]. The mechanisms for this variation remain unclear but could relate to specific sexual practices.

TRANSMISSION — Human herpesvirus-8 (HHV-8) is primarily transmitted by saliva but can also be transmitted via organ donation or rarely, via blood transfusion.

Salivary transmission – The mode of transmission of HHV-8 is known to be primarily via saliva. However, the mechanism by which salivary HHV-8 transmission occurs differs by epidemiologic context [40].

In endemic areas, including Africa, vertical (mother-to-child) salivary transmission is thought to be the major method [41-43]. In these areas, HHV-8 seropositivity is relatively uncommon in infants, and there is an age-dependent increase in HHV-8 seroprevalence during childhood, demonstrating that neither in utero nor vertical transmission by breast feeding are major contributors but showing that other routes of vertical and household transmission are likely to be important [43]. Children are more likely to be infected if they reside with an affected or seropositive mother or other family member [44]. Maternal pre-mastication of food is a likely contributor to this process, though other behaviors are also the subject of exploration [43]. Supporting the salivary theory of transmission, studies of salivary secretions show detectable HHV-8 DNA in a proportion of individuals with infection, often on an intermittent basis [40]. Increased oral HHV-8 DNA detection, with implications for onward transmission, is seen with untreated HIV infection, malaria parasitemia, and perhaps other infections including herpes simplex [45,46]

In contrast, vertical salivary transmission is uncommon outside endemic areas. In these settings, establishing the mechanisms by which elevated HHV-8 seroprevalence rates were established and maintained in key risk groups including men who have sex with men (MSM) has been challenging [35]. The age pattern of increases in prevalence is relatively late in nonendemic areas, occurring well after sexual debut (in contrast for example to salivary transmission of EBV around sexual debut in these settings) [35,47]. The elevated prevalence in MSM specifically and in people with more sexual partners generally is suggestive of routes of transmission related to sexual activity, potentially via saliva during intercourse [37,40,47-49]. Seminal fluid appears to be much less important in transmission, based on studies of HHV-8 detection in semen of individuals with infection [50,51].

Organ donation – Transmission of HHV-8 of donor origin to recipients of solid organ transplants has been reported and may be more common than is generally recognized clinically [52]. Such transmission likely arises from HHV-8-infected mononuclear cells included in an organ transplanted to a seronegative recipient [53,54]. In one prospective study of 217 HHV-8 seronegative patients receiving transplants from seropositive organ donors, approximately 30 percent of recipients seroconverted to HHV-8 [55]. While most seroconversion in this setting is asymptomatic, clinical sequelae can occur, influenced in part by immunosuppression following transplantation. Severe inflammatory symptoms and fatal visceral Kaposi sarcoma (KS) with HHV-8-associated lymphoproliferation have been reported in seronegative recipients developing primary HHV-8 infection in this context [56,57] (see "Malignancy after solid organ transplantation", section on 'Kaposi sarcoma'). Consideration should be given to monitoring HHV-8 serostatus in solid organ donors and recipients and monitoring for clinical evidence of primary infection.

Blood transfusion – HHV-8 transmission during transfusion of blood and blood products is possible, via HHV-8-infected mononuclear cells included in donated and transfused blood products. HHV-8 is generally not included in infectious diseases screening of donors and donated blood products. Despite this, however, in practice such transmission appears rare [58]. The lack of observed transmission likely reflects several factors. These include the impact of donor screening and laboratory testing for other infectious diseases, both of which likely also reduce the rates of HHV-8 infection in accepted blood donors, along with processing leucodepletion of products prior to transfusion. In addition, even in infected donors, rates of detectable viremia in peripheral blood (which would establish the conditions for onward transmission via donated blood) are extremely low [59]. In resource-limited settings where HHV-8 is endemic, donor screening more limited, and leucodepletion less common, HHV-8 transmission via transfusion may be more clinically important [58].

ASSOCIATED CLINICAL SYNDROMES — Aside from a generally asymptomatic primary infection, human herpesvirus-8 (HHV-8) is associated with multiple tumor syndromes and a clinical syndrome called Kaposi sarcoma inflammatory cytokine syndrome (KICS). Primary HHV-8 infection is described in detail below and the details of the other associated clinical syndromes are discussed in detail separately.

Primary infection — In most cases, primary infection with HHV-8 is either asymptomatic or minimally symptomatic. However, symptomatic primary infection syndrome has been described in immunocompetent children, immunocompetent adults, and immunocompromised individuals.

Children – In some immunocompetent children, primary HHV-8 infection may be associated with fever and a maculopapular rash [60]. In a small series, five of six children with evidence of primary infection and seroconversion developed a centripetal maculopapular rash, appearing on the face and spreading gradually to the trunk and extremities. The median duration of the rash was six days (range three to eight days), and the median duration of fever was 10 days (range 2 to 14 days).

Immunocompetent adults – In immunocompetent adults, studies in men who have sex with men (MSM) without HIV describe transient lymphadenopathy in association with HHV-8 seroconversion [36,61]. In a minority of cases, the lymphadenopathy was associated with a mild systemic illness including fatigue, diarrhea, and localized rash [36].

Immunocompromised individuals – In immunocompromised adults, including solid organ transplant recipients and individuals with HIV, case reports describe significant systemic symptoms with primary infection. Features include fever, splenomegaly, lymphoid hyperplasia, pancytopenia, and in some cases rapid-onset KS [62]. These features resemble the inflammatory symptoms seen with HHV-8-associated multicentric Castleman disease (MCD) and with KICS, but the great majority of cases were transient and self-limited [6,7].

Kaposi sarcoma — Kaposi sarcoma (KS) is a multifocal angioproliferative tumor with four epidemiologic forms: Classic KS, endemic or African KS, AIDS-related KS, and organ transplant-associated KS (table 1). Regardless of these epidemiologic categories, the pathology of KS is similar, and with some exception, the clinical course is similar as well. KS usually manifests as red, purple, or brown papules or plaques on the skin or mucous membranes [63] (picture 1 and picture 2 and picture 3). Lesions may occur at any site, but there is a predilection for the extremities (picture 4 and picture 5), ears, nose (picture 6), and palate (picture 7) for reasons which remain unclear. KS involvement of visceral organs such as the lungs or gastrointestinal tract and effusions in serous body cavities are other manifestations of advanced disease and may be life threatening [64,65]. (See "AIDS-related Kaposi sarcoma: Clinical manifestations and diagnosis" and "Classic Kaposi sarcoma: Clinical features, staging, diagnosis, and treatment" and "Malignancy after solid organ transplantation", section on 'Kaposi sarcoma'.)

Multicentric Castleman disease — HHV-8 is the etiologic agent of a plasmablastic form of MCD that is most common in the setting of HIV but can also arise in transplant recipients and in other patients without HIV [66]. HHV-8 associated MCD is a polyclonal B cell disorder characterized clinically by intermittent flares of inflammatory symptoms and signs, including fevers, night sweats, fatigue, and cachexia, and edema, together with lymphadenopathy and hepatosplenomegaly. Common laboratory abnormalities include anemia, thrombocytopenia, hypoalbuminemia, hyponatremia, and elevated inflammatory markers, most notably C-reactive protein (CRP) [5,67]. (See "HHV-8/KSHV-associated multicentric Castleman disease".)

HHV-8 (Kaposi sarcoma) inflammatory cytokine syndrome (KICS) — HHV-8/Kaposi sarcoma inflammatory cytokine syndrome (KICS) is a syndrome characterized by severe inflammatory symptoms and elevated HHV-8 viremia [6,7]. The clinical manifestations are similar to those of MCD, and include fevers, night sweats, fatigue, and cachexia, and edema. Hepatosplenomegaly is common whereas lymphadenopathy is not [7]. Laboratory abnormalities include anemia, thrombocytopenia, hypoalbuminemia, hyponatremia, and elevated inflammatory markers including C-reactive protein. (See "AIDS-related Kaposi sarcoma: Clinical manifestations and diagnosis", section on 'Inflammatory cytokine syndrome'.)

Primary effusion lymphoma (PEL) — PEL is a rare variant of B-cell non-Hodgkin lymphoma (NHL) notable for its unusual presentation and aggressive clinical course [68,69]. The great majority of reported cases occur in people with HIV, although it may also be seen following solid organ transplantation, in older adults, and in chronic hepatitis C virus infection [70]. PEL usually presents as a lymphomatous effusion in serous body cavities, with pleural involvement seen in 60 to 90 percent of patients, followed by involvement of other body cavity membranes, including peritoneal (30 to 60 percent), pericardial (up to 30 percent), joint spaces, and rarely meninges [68,69,71]. (See "Primary effusion lymphoma".)

DIAGNOSIS

When to suspect HHV-8 infection — Human herpesvirus-8 (HHV-8) infection should be suspected when an immunocompromised patient presents with symptoms or signs that are consistent with one of the HHV-8 associated clinical tumor syndromes (Kaposi sarcoma, multicentric Castleman disease, or primary effusion lymphoma).

Since primary HHV-8 infection is transient and self-limiting, even in immunocompromised individuals, diagnosis and treatment is not necessary.

There is no clinical role for screening asymptomatic individuals for HHV-8 infection.

Diagnostic tests — Available tests to detect HHV-8 include immunohistochemistry on body tissue/fluid, blood polymerase chain reaction (PCR), and serology. HHV-8 associated tumors are typically diagnosed by immunohistochemistry of body tissue samples or cells from body cavity effusions. In contrast, Kaposi sarcoma inflammatory cytokine syndrome (KICS) is typically diagnosed by detecting HHV-8 viremia by PCR in peripheral blood.

There is no US Food and Drug Administration (FDA)-approved diagnostic test for clinical purposes, such as documentation of acute infection. As a result, important clinical challenges exist related to the lack of a gold-standard confirmatory test for HHV-8 infection in subjects without a documented HHV-8-associated malignancy or detectable HHV-8 in the blood or saliva.

Serology — Available serologic tests are not FDA approved and are not suitable for establishing the presence of HHV-8 at an individual level. Although assessment of HHV-8 prevalence in populations is based on serologic antibody testing, these assays are usually not sufficiently robust for clinical use to ascertain an individual's infection status (eg, to document acute or chronic infection), as discussed below [72]. Current HHV-8 serologic tests use either immunofluorescent assays (IFAs) or enzyme-linked immunoassays (ELISAs) against one or more HHV-8-encoded latent and/or lytic proteins: one lytic (K8.1) and latent (LANA) protein is commonly used to increase sensitivity while preserving specificity [29,73,74]. The sensitivity of these assays is variable , while the specificity is generally greater than 95 percent [72].

Polymerase chain reaction (PCR) — There are several different PCR assays that employ primers unique for HHV-8. HHV-8 DNA can be identified using PCR in virtually all biopsy samples of Kaposi sarcoma (KS) and HHV-8-associated multicentric Castleman disease (MCD) and in the peripheral blood of individuals with KICS and MCD during active disease flares [5,7,75,76]. Clinical applications of PCR for HHV-8 disease are predominantly to support a diagnosis of HHV-8-associated MCD or KICS in those with characteristic clinical features.

HHV-8 quantification in plasma or peripheral blood mononuclear cells by PCR may be a useful means for diagnosing an active flare of HHV8-associated MCD or KICS, and in following response to treatment [7,75]. HHV-8 DNA can also be detected by PCR in tumor cells within the pleural fluid in individuals with PEL, but this is less clinically useful in diagnosis or monitoring therapeutic response [69].

HHV-8 viremia can also be detected in asymptomatic individuals. The prevalence of viremia in persons asymptomatically infected with HHV-8 ranges from 4 to 20 percent [59]; thus PCR testing should not generally be done in patients without symptoms concerning for KICS. A positive HHV-8 PCR in the blood of a patient who does not demonstrate symptoms of KICS or HHV-8 associated tumors is not indicative of active disease.

Most PCR-based methods do not distinguish between latent and lytically replicating virus. As HHV-8 is often present sporadically in tissue, detection of low-level viremia is frequent and usually of no clinical significance.

HHV-8 viremia may be prognostic in other settings but is not yet in common clinical use for this purpose. For example, in sub-Saharan Africa where the detection of HHV-8 in the peripheral blood is common among individuals with KS, the level of HHV-8 DNA in the plasma appears useful in both predicting survival and response to treatment after a diagnosis of KS [77].

TREATMENT — Several antiviral agents such as ganciclovir, foscarnet, and cidofovir inhibit human herpesvirus-8 (HHV-8) replication in vitro [78,79]. However, direct antiviral therapy of HHV-8 infection in patients has a very limited role. Primary HHV-8 infection is usually asymptomatic and if symptomatic, self-limited without treatment. Antiviral agents have not been shown to have activity in treating established Kaposi sarcoma (KS), although ganciclovir, when used to treat cytomegalovirus (CMV) retinitis, did lower the risk of KS in patients with AIDS in a study conducted in the pre-antiretroviral therapy (ART) era [80]. Case series and small studies have demonstrated that antiviral agents including ganciclovir alone or in combination with other agents can have activity in HHV-8 associated multicentric Castleman disease (MCD) [81,82], but its use has almost entirely been supplanted by immunomodulating therapies, most notably rituximab [66]. (See "AIDS-related Kaposi sarcoma: Staging and treatment" and "Classic Kaposi sarcoma: Clinical features, staging, diagnosis, and treatment", section on 'Treatment' and "HHV-8/KSHV-associated multicentric Castleman disease", section on 'Treatment' and "Primary effusion lymphoma", section on 'Management'.)

HHV-8 infection cannot be eradicated although long-term remissions of the associated clinical syndromes are possible with reduction in immunosuppression in immunocompromised patients [83-86].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Opportunistic infections in adults with HIV".)

SUMMARY AND RECOMMENDATIONS

Virology and epidemiology – Human herpesvirus-8 (HHV-8) is a large (165 kB) double-stranded DNA gamma (γ2) herpesvirus which is endemic worldwide (especially in sub-Saharan Africa and parts of the Mediterranean) and more common in certain behavioral groups such as men who have sex with men (MSM). It is classified as a group 1 biological carcinogenic agent. (See 'Virology' above and 'Epidemiology' above.)

Transmission – HHV-8 is primarily transmitted by saliva during childhood but can also be transmitted via organ donation or rarely, via blood transfusion. (See 'Transmission' above.)

Associated clinical syndromes – Most individuals infected with HHV-8 will experience no clinical consequences. However, in some cases, especially in immunocompromised individuals, HHV-8 can cause Kaposi sarcoma (KS), one form of the inflammatory lymphoproliferative disorder multicentric Castleman disease (MCD; HHV-8-associated MCD), primary effusion lymphoma (PEL), and/or the inflammatory syndrome known as the HHV-8 inflammatory cytokine syndrome (KICS). (See 'Associated clinical syndromes' above.)

Diagnosis

HHV-8 infection should be suspected when an immunocompromised patient presents with symptoms that are consistent with one of the HHV-8 associated clinical syndromes (Kaposi sarcoma, multicentric Castleman disease, primary effusion lymphoma, or KICS). (See 'When to suspect HHV-8 infection' above.)

Diagnosis of primary HHV-8 infection or screening of asymptomatic individuals for HHV-8 infection is not indicated. (See 'When to suspect HHV-8 infection' above.)

HHV-8 associated tumors are typically diagnosed by immunohistochemistry of body tissue samples or cells from body cavity effusions. (See 'Diagnostic tests' above.)

Treatment – There is no role for direct antiviral therapy of HHV-8 infection as most primary HHV-8 infections are asymptomatic and/or self-limited. Treatment of HHV-8 associated clinical syndromes are discussed separately. (See 'Treatment' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Corey Casper, MD, MPH, who contributed to earlier versions of this topic review.

  1. Chang Y, Cesarman E, Pessin MS, et al. Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi's sarcoma. Science 1994; 266:1865.
  2. International Agency for Research on Cancer. A review of human carcinogens. In: Biological agents/IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, World Health Organization, Lyon, France 2009. Vol 100B. https://monographs.iarc.who.int/wp-content/uploads/2018/06/mono100B.pdf.
  3. Bouvard V, Baan R, Straif K, et al. A review of human carcinogens--Part B: biological agents. Lancet Oncol 2009; 10:321.
  4. Soulier J, Grollet L, Oksenhendler E, et al. Kaposi's sarcoma-associated herpesvirus-like DNA sequences in multicentric Castleman's disease. Blood 1995; 86:1276.
  5. Oksenhendler E, Duarte M, Soulier J, et al. Multicentric Castleman's disease in HIV infection: a clinical and pathological study of 20 patients. AIDS 1996; 10:61.
  6. Uldrick TS, Wang V, O'Mahony D, et al. An interleukin-6-related systemic inflammatory syndrome in patients co-infected with Kaposi sarcoma-associated herpesvirus and HIV but without Multicentric Castleman disease. Clin Infect Dis 2010; 51:350.
  7. Polizzotto MN, Uldrick TS, Wyvill KM, et al. Clinical Features and Outcomes of Patients With Symptomatic Kaposi Sarcoma Herpesvirus (KSHV)-associated Inflammation: Prospective Characterization of KSHV Inflammatory Cytokine Syndrome (KICS). Clin Infect Dis 2016; 62:730.
  8. Teruya-Feldstein J, Zauber P, Setsuda JE, et al. Expression of human herpesvirus-8 oncogene and cytokine homologues in an HIV-seronegative patient with multicentric Castleman's disease and primary effusion lymphoma. Lab Invest 1998; 78:1637.
  9. Byun M, Ma CS, Akçay A, et al. Inherited human OX40 deficiency underlying classic Kaposi sarcoma of childhood. J Exp Med 2013; 210:1743.
  10. Grossman WJ, Radhi M, Schauer D, et al. Development of hemophagocytic lymphohistiocytosis in triplets infected with HHV-8. Blood 2005; 106:1203.
  11. Staskus KA, Sun R, Miller G, et al. Cellular tropism and viral interleukin-6 expression distinguish human herpesvirus 8 involvement in Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. J Virol 1999; 73:4181.
  12. Moore PS, Chang Y. Kaposi's sarcoma-associated herpesvirus-encoded oncogenes and oncogenesis. J Natl Cancer Inst Monogr 1998; :65.
  13. Wen KW, Damania B. Kaposi sarcoma-associated herpesvirus (KSHV): molecular biology and oncogenesis. Cancer Lett 2010; 289:140.
  14. Moore PS, Boshoff C, Weiss RA, Chang Y. Molecular mimicry of human cytokine and cytokine response pathway genes by KSHV. Science 1996; 274:1739.
  15. Boshoff C, Endo Y, Collins PD, et al. Angiogenic and HIV-inhibitory functions of KSHV-encoded chemokines. Science 1997; 278:290.
  16. Davis DA, Rinderknecht AS, Zoeteweij JP, et al. Hypoxia induces lytic replication of Kaposi sarcoma-associated herpesvirus. Blood 2001; 97:3244.
  17. Li X, Feng J, Sun R. Oxidative stress induces reactivation of Kaposi's sarcoma-associated herpesvirus and death of primary effusion lymphoma cells. J Virol 2011; 85:715.
  18. Gregory SM, West JA, Dillon PJ, et al. Toll-like receptor signaling controls reactivation of KSHV from latency. Proc Natl Acad Sci U S A 2009; 106:11725.
  19. Zeng Y, Zhang X, Huang Z, et al. Intracellular Tat of human immunodeficiency virus type 1 activates lytic cycle replication of Kaposi's sarcoma-associated herpesvirus: role of JAK/STAT signaling. J Virol 2007; 81:2401.
  20. Labo N, Miley W, Marshall V, et al. Heterogeneity and breadth of host antibody response to KSHV infection demonstrated by systematic analysis of the KSHV proteome. PLoS Pathog 2014; 10:e1004046.
  21. de Martel C, Georges D, Bray F, et al. Global burden of cancer attributable to infections in 2018: a worldwide incidence analysis. Lancet Glob Health 2020; 8:e180.
  22. Gao SJ, Kingsley L, Li M, et al. KSHV antibodies among Americans, Italians and Ugandans with and without Kaposi's sarcoma. Nat Med 1996; 2:925.
  23. de Sanjose S, Mbisa G, Perez-Alvarez S, et al. Geographic variation in the prevalence of Kaposi sarcoma-associated herpesvirus and risk factors for transmission. J Infect Dis 2009; 199:1449.
  24. Sitas F, Carrara H, Beral V, et al. Antibodies against human herpesvirus 8 in black South African patients with cancer. N Engl J Med 1999; 340:1863.
  25. Biggar RJ, Rosenberg PS, Coté T. Kaposi's sarcoma and non-Hodgkin's lymphoma following the diagnosis of AIDS. Multistate AIDS/Cancer Match Study Group. Int J Cancer 1996; 68:754.
  26. Biggar RJ, Chaturvedi AK, Goedert JJ, et al. AIDS-related cancer and severity of immunosuppression in persons with AIDS. J Natl Cancer Inst 2007; 99:962.
  27. Shiels MS, Pfeiffer RM, Hall HI, et al. Proportions of Kaposi sarcoma, selected non-Hodgkin lymphomas, and cervical cancer in the United States occurring in persons with AIDS, 1980-2007. JAMA 2011; 305:1450.
  28. Lennette ET, Blackbourn DJ, Levy JA. Antibodies to human herpesvirus type 8 in the general population and in Kaposi's sarcoma patients. Lancet 1996; 348:858.
  29. Simpson GR, Schulz TF, Whitby D, et al. Prevalence of Kaposi's sarcoma associated herpesvirus infection measured by antibodies to recombinant capsid protein and latent immunofluorescence antigen. Lancet 1996; 348:1133.
  30. Whitby D, Luppi M, Barozzi P, et al. Human herpesvirus 8 seroprevalence in blood donors and lymphoma patients from different regions of Italy. J Natl Cancer Inst 1998; 90:395.
  31. Biggar RJ, Whitby D, Marshall V, et al. Human herpesvirus 8 in Brazilian Amerindians: a hyperendemic population with a new subtype. J Infect Dis 2000; 181:1562.
  32. Rezza G, Danaya RT, Wagner TM, et al. Human herpesvirus-8 and other viral infections, Papua New Guinea. Emerg Infect Dis 2001; 7:893.
  33. Brown EE, Fallin D, Ruczinski I, et al. Associations of classic Kaposi sarcoma with common variants in genes that modulate host immunity. Cancer Epidemiol Biomarkers Prev 2006; 15:926.
  34. Smith NA, Sabin CA, Gopal R, et al. Serologic evidence of human herpesvirus 8 transmission by homosexual but not heterosexual sex. J Infect Dis 1999; 180:600.
  35. Martin JN, Ganem DE, Osmond DH, et al. Sexual transmission and the natural history of human herpesvirus 8 infection. N Engl J Med 1998; 338:948.
  36. Casper C, Wald A, Pauk J, et al. Correlates of prevalent and incident Kaposi's sarcoma-associated herpesvirus infection in men who have sex with men. J Infect Dis 2002; 185:990.
  37. Gras J, Helary M, Carette D, et al. Prevalence, Risk factors, and Shedding of Human Herpes Virus-8 among Men Having Sex With Men Enrolled in a Pre-exposure Prophylaxis Study. Clin Infect Dis 2023.
  38. Knights SM, Salyards M, Kendall N, et al. High Seroprevalence of Kaposi Sarcoma-Associated Herpesvirus in Men Who Have Sex With Men With HIV in the Southern United States. Open Forum Infect Dis 2023; 10:ofad160.
  39. Giuliani M, Cordiali-Fei P, Castilletti C, et al. Incidence of human herpesvirus 8 (HHV-8) infection among HIV-uninfected individuals at high risk for sexually transmitted infections. BMC Infect Dis 2007; 7:143.
  40. Pauk J, Huang ML, Brodie SJ, et al. Mucosal shedding of human herpesvirus 8 in men. N Engl J Med 2000; 343:1369.
  41. Taylor MM, Chohan B, Lavreys L, et al. Shedding of human herpesvirus 8 in oral and genital secretions from HIV-1-seropositive and -seronegative Kenyan women. J Infect Dis 2004; 190:484.
  42. Mbulaiteye SM, Atkinson JO, Whitby D, et al. Risk factors for human herpesvirus 8 seropositivity in the AIDS Cancer Cohort Study. J Clin Virol 2006; 35:442.
  43. Butler LM, Dorsey G, Hladik W, et al. Kaposi sarcoma-associated herpesvirus (KSHV) seroprevalence in population-based samples of African children: evidence for at least 2 patterns of KSHV transmission. J Infect Dis 2009; 200:430.
  44. Mbulaiteye SM, Biggar RJ, Pfeiffer RM, et al. Water, socioeconomic factors, and human herpesvirus 8 infection in Ugandan children and their mothers. J Acquir Immune Defic Syndr 2005; 38:474.
  45. Wakeham K, Webb EL, Sebina I, et al. Risk factors for seropositivity to Kaposi sarcoma-associated herpesvirus among children in Uganda. J Acquir Immune Defic Syndr 2013; 63:228.
  46. Engels EA, Atkinson JO, Graubard BI, et al. Risk factors for human herpesvirus 8 infection among adults in the United States and evidence for sexual transmission. J Infect Dis 2007; 196:199.
  47. Dukers NH, Renwick N, Prins M, et al. Risk factors for human herpesvirus 8 seropositivity and seroconversion in a cohort of homosexual men. Am J Epidemiol 2000; 151:213.
  48. Kedes DH, Operskalski E, Busch M, et al. The seroepidemiology of human herpesvirus 8 (Kaposi's sarcoma-associated herpesvirus): distribution of infection in KS risk groups and evidence for sexual transmission. Nat Med 1996; 2:918.
  49. Salyards M, Nijhawan AE, Kuo J, et al. Prevalence, Incidence, and Predictors of Kaposi Sarcoma-associated Herpesvirus Infection Among Young Men Who Have Sex with Men in the Southern United States. J Infect Dis 2023.
  50. Ambroziak JA, Blackbourn DJ, Herndier BG, et al. Herpes-like sequences in HIV-infected and uninfected Kaposi's sarcoma patients. Science 1995; 268:582.
  51. Monini P, de Lellis L, Fabris M, et al. Kaposi's sarcoma-associated herpesvirus DNA sequences in prostate tissue and human semen. N Engl J Med 1996; 334:1168.
  52. Regamey N, Tamm M, Wernli M, et al. Transmission of human herpesvirus 8 infection from renal-transplant donors to recipients. N Engl J Med 1998; 339:1358.
  53. Barozzi P, Luppi M, Facchetti F, et al. Post-transplant Kaposi sarcoma originates from the seeding of donor-derived progenitors. Nat Med 2003; 9:554.
  54. Dudderidge TJ, Khalifa M, Jeffery R, et al. Donor-derived human herpes virus 8-related Kaposi's sarcoma in renal allograft ureter. Transpl Infect Dis 2008; 10:221.
  55. Lebbe C, Porcher R, Marcelin AG, et al. Human herpesvirus 8 (HHV8) transmission and related morbidity in organ recipients. Am J Transplant 2013; 13:207.
  56. Francès C, Marcelin AG, Legendre Ch, et al. The impact of preexisting or acquired Kaposi sarcoma herpesvirus infection in kidney transplant recipients on morbidity and survival. Am J Transplant 2009; 9:2580.
  57. Pietrosi G, Vizzini G, Pipitone L, et al. Primary and reactivated HHV8 infection and disease after liver transplantation: a prospective study. Am J Transplant 2011; 11:2715.
  58. Hladik W, Dollard SC, Mermin J, et al. Transmission of human herpesvirus 8 by blood transfusion. N Engl J Med 2006; 355:1331.
  59. Kleinman S, King MR, Busch MP, et al. The National Heart, Lung, and Blood Institute retrovirus epidemiology donor studies (Retrovirus Epidemiology Donor Study and Retrovirus Epidemiology Donor Study-II): twenty years of research to advance blood product safety and availability. Transfus Med Rev 2012; 26:281.
  60. Andreoni M, Sarmati L, Nicastri E, et al. Primary human herpesvirus 8 infection in immunocompetent children. JAMA 2002; 287:1295.
  61. Wang QJ, Jenkins FJ, Jacobson LP, et al. Primary human herpesvirus 8 infection generates a broadly specific CD8(+) T-cell response to viral lytic cycle proteins. Blood 2001; 97:2366.
  62. Oksenhendler E, Cazals-Hatem D, Schulz TF, et al. Transient angiolymphoid hyperplasia and Kaposi's sarcoma after primary infection with human herpesvirus 8 in a patient with human immunodeficiency virus infection. N Engl J Med 1998; 338:1585.
  63. Martró E, Esteve A, Schulz TF, et al. Risk factors for human Herpesvirus 8 infection and AIDS-associated Kaposi's sarcoma among men who have sex with men in a European multicentre study. Int J Cancer 2007; 120:1129.
  64. Cesarman E, Damania B, Krown SE, et al. Kaposi sarcoma. Nat Rev Dis Primers 2019; 5:9.
  65. Davidson A, Wainwright RD, Stones DK, et al. Malignancies in South African children with HIV. J Pediatr Hematol Oncol 2014; 36:111.
  66. Carbone A, Borok M, Damania B, et al. Castleman disease. Nat Rev Dis Primers 2021; 7:84.
  67. Mylona EE, Baraboutis IG, Lekakis LJ, et al. Multicentric Castleman's disease in HIV infection: a systematic review of the literature. AIDS Rev 2008; 10:25.
  68. Boulanger E, Gérard L, Gabarre J, et al. Prognostic factors and outcome of human herpesvirus 8-associated primary effusion lymphoma in patients with AIDS. J Clin Oncol 2005; 23:4372.
  69. Lurain K, Polizzotto MN, Aleman K, et al. Viral, immunologic, and clinical features of primary effusion lymphoma. Blood 2019; 133:1753.
  70. Engels EA, Pfeiffer RM, Fraumeni JF Jr, et al. Spectrum of cancer risk among US solid organ transplant recipients. JAMA 2011; 306:1891.
  71. Nador RG, Cesarman E, Chadburn A, et al. Primary effusion lymphoma: a distinct clinicopathologic entity associated with the Kaposi's sarcoma-associated herpes virus. Blood 1996; 88:645.
  72. Engels EA, Whitby D, Goebel PB, et al. Identifying human herpesvirus 8 infection: performance characteristics of serologic assays. J Acquir Immune Defic Syndr 2000; 23:346.
  73. Gao SJ, Kingsley L, Hoover DR, et al. Seroconversion to antibodies against Kaposi's sarcoma-associated herpesvirus-related latent nuclear antigens before the development of Kaposi's sarcoma. N Engl J Med 1996; 335:233.
  74. Mbisa GL, Miley W, Gamache CJ, et al. Detection of antibodies to Kaposi's sarcoma-associated herpesvirus: a new approach using K8.1 ELISA and a newly developed recombinant LANA ELISA. J Immunol Methods 2010; 356:39.
  75. Polizzotto MN, Uldrick TS, Wang V, et al. Human and viral interleukin-6 and other cytokines in Kaposi sarcoma herpesvirus-associated multicentric Castleman disease. Blood 2013; 122:4189.
  76. Stebbing J, Adams C, Sanitt A, et al. Plasma HHV8 DNA predicts relapse in individuals with HIV-associated multicentric Castleman disease. Blood 2011; 118:271.
  77. Tedeschi R, Enbom M, Bidoli E, et al. Viral load of human herpesvirus 8 in peripheral blood of human immunodeficiency virus-infected patients with Kaposi's sarcoma. J Clin Microbiol 2001; 39:4269.
  78. Kedes DH, Ganem D. Sensitivity of Kaposi's sarcoma-associated herpesvirus replication to antiviral drugs. Implications for potential therapy. J Clin Invest 1997; 99:2082.
  79. Casper C, Krantz EM, Corey L, et al. Valganciclovir for suppression of human herpesvirus-8 replication: a randomized, double-blind, placebo-controlled, crossover trial. J Infect Dis 2008; 198:23.
  80. Martin DF, Kuppermann BD, Wolitz RA, et al. Oral ganciclovir for patients with cytomegalovirus retinitis treated with a ganciclovir implant. Roche Ganciclovir Study Group. N Engl J Med 1999; 340:1063.
  81. Casper C, Nichols WG, Huang ML, et al. Remission of HHV-8 and HIV-associated multicentric Castleman disease with ganciclovir treatment. Blood 2004; 103:1632.
  82. Uldrick TS, Polizzotto MN, Aleman K, et al. High-dose zidovudine plus valganciclovir for Kaposi sarcoma herpesvirus-associated multicentric Castleman disease: a pilot study of virus-activated cytotoxic therapy. Blood 2011; 117:6977.
  83. Bower M, Fox P, Fife K, et al. Highly active anti-retroviral therapy (HAART) prolongs time to treatment failure in Kaposi's sarcoma. AIDS 1999; 13:2105.
  84. Martinez V, Caumes E, Gambotti L, et al. Remission from Kaposi's sarcoma on HAART is associated with suppression of HIV replication and is independent of protease inhibitor therapy. Br J Cancer 2006; 94:1000.
  85. Stallone G, Infante B, Grandaliano G, et al. Kaposi's sarcoma and mTOR: a crossroad between viral infection neoangiogenesis and immunosuppression. Transpl Int 2008; 21:825.
  86. Maskew M, Fox MP, van Cutsem G, et al. Treatment response and mortality among patients starting antiretroviral therapy with and without Kaposi sarcoma: a cohort study. PLoS One 2013; 8:e64392.
Topic 8319 Version 21.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟