Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 May 1.
Published in final edited form as: J Med Virol. 2023 May;95(5):e28773. doi: 10.1002/jmv.28773

Today’s Kaposi Sarcoma is not the same as it was 40 years ago, or is it?

Blossom Damania 1,*, Dirk P Dittmer 1,*
PMCID: PMC10266714  NIHMSID: NIHMS1902915  PMID: 37212317

Abstract

This review will provide an overview of the notion that Kaposi Sarcoma (KS) is a disease that manifests under diverse and divergent circumstances. We begin with a historical introduction of KS and KS-associated herpesvirus (KSHV), highlight the diversity of clinical presentations of KS, summarize what we know about the cell of origin for this tumor, explore KSHV viral load as a potential biomarker for acute KSHV infections and KS-associated complications, and discuss immune modulators that impact KSHV infection, KSHV persistence and KS disease.

Keywords: Kaposi Sarcoma, Herpesvirus, KSHV

Introduction

In 1981 a cluster of Kaposi Sarcoma (KS) diagnoses in New York and San Francisco heralded the beginning of the Acquired Immune Deficiency Syndrome (AIDS) pandemic 1. KS was a clinical marker of AIDS before either human immunodeficiency virus (HIV), or KS-associated herpesvirus (KSHV), were discovered. Multifocal lesions, sometimes flat, sometimes nodular, indicated a severe loss of adaptive immune control. At that time, in HIV-uncontrolled AIDS patients, KS lesions occurred at CD4 counts below 200 cells per μl. This manifestation of KSHV infection is commonly referred to as AIDS-KS. AIDS-KS has skin and internal manifestations (Figure 1ALung lesion picture). AIDS-KS patients have or had concurrent infections of KSHV with other microorganisms, such as viruses, fungi, or bacteria.

Figure 1.

Figure 1

A. Picture of KS lesions in the lung. Arrows indicate a nodular lesion (N) and a flat lesion (F). B. Distribution of KSHV plasma viral loads (DNA genome copies/ml) in a cohort of 29 PLWH on successful cART presenting with KS requiring cancer therapy (data from 13).

Today, 40 years later, late-stage AIDS cases are rarer, but KS remains among the leading cancers in male people living with HIV (PLWH), including those who never developed AIDS and successfully suppressed HIV viral loads 24. This begs the question: is the KS we treat today the same as it was 40 years ago? Is it the same as almost 200 years ago when Moritz Kaposi discovered this disease in HIV-negative, elderly men?

Moritz Kaposi first described KS in 1884 in Vienna, Austria (reviewed in 5). At the time, Vienna was the most cosmopolitan city on earth. Here, travelers and migrants from across the realm of the Habsburg empire converged. Since 1883 the Orient Express connected Vienna with Constantinople and from there to the Levant, i.e., the eastern Mediterranean. Moritz Kaposi was the director of the skin clinic at the Vienna General Hospital. He was among the first to describe xeroderma pigmentosum. At that time, the Vienna General Hospital was the site of the famed Viennese Medical School, where some years earlier, Ignaz Semmelweis formulated basic ideas of hygiene. In broad strokes, one can compare Vienna General Hospital in 1880 with San Francisco General Hospital in 1980: both were centers of clinical excellence where physicians conducted discovery science.

The first case of Sub-Saharan KS was noted in 1960 6, before the emergence of HIV in the region and shortly after Dennis Burkitt described Burkitt lymphoma (BL) in the same country. BL, of course, is famously associated with Epstein-Barr Virus (EBV) but not with HIV. Once the viral association between EBV and BL was established, the idea of tumor-causing viruses gained acceptance. It was reinforced by the discovery of the human papillomavirus (HPV) -- cervical cancer association. Since then, the possibility of an infectious etiology for human cancers has been on physicians’ and epidemiologists’ minds. The emergence of HIV-associated cancers suggested an infectious agent as the cause of KS. It led to the discovery by Chang and Moore of Kaposi Sarcoma-associated herpesvirus (KSHV) 7 in 1994. Once the viral etiology of KS was revealed, research began in earnest, and our understanding of the pathobiology of this cancer grew exponentially. To date, PubMed records 17,108 entries under the keyword “Kaposi sarcoma.”

KS has diverse manifestations but a unified etiology: KSHV.

A recent estimate counts non-Hodgkin lymphoma (NHL), KS, HPV-associated anal cancer, and lung cancer as the most significant cancer contributors to years of life lost among PLWH in the US; these diseases are concentrated mainly in black PLWH, men who have sex with men (MSM), and the 40–59 year age bracket 8. The risk of developing KS in PLWH is elevated several hundred-fold compared with the general population 9. The human oncogenic herpesvirus KSHV is the underlying etiological agent for clinical manifestations recorded at the turn of the 19th century Vienna, in AIDS patients in 1981 New York and San Francisco, children in eastern Africa, and adults in Xinjiang province in China, today 1,10,11.

In KSHV-endemic areas, KSHV acquisition precedes symptomatic disease by many years (the exception being pediatric KS). In 1980s AIDS patients, KSHV seroconversion preceded KS disease by just ~33 months 12. This may be an anomaly, however, brought about by the simultaneous introduction of both HIV and KSHV into a population that experienced high transmission rates for both viruses. It is important to reemphasize that even in PLWH, KS can develop without detectable HIV and near-normal CD4 counts 2,3. We do not have a specific name for this subtype of KS but would suggest that it not be called AIDS-KS because the patients do not have AIDS, only KS. For this review, we refer to it as HIV-KS.

The unifying principle of all KS is exposure to KSHV. KSHV infection is either accompanied or followed by transient immune deficiency that predisposes a person to KS. If there is no virus encounter, then there is no cancer predisposition. Thus, KSHV partially fulfills Koch’s first postulate. Not every KSHV-infected person develops KS, however. Instead, a fixed probability exists that a person infected with KSHV will develop clinically apparent disease. This holds for all the human oncogenic viruses, KSHV, EBV, HPV, human T-lymphotropic virus (HTLV-1), hepatitis C virus (HCV), and Merkel Cell Polyomavirus (MCPyV). The immune status and other factors influence the probability of conversion from infection to clinical disease.

KSHV is present at varying copy numbers in the blood of people who develop KS. For example, Figure 1B shows viral load data from a cohort of PLWH on stable combination antiretroviral therapy (cART) requiring anti-cancer therapy for their KS 13. While most patients with clinically apparent disease had detectable KSHV genomes in blood, KSHV viremia with >1,000 copies per ml was rare, even for patients with multiple KS lesions. This is consistent with transcriptional profiling data that show most KS lesions harbor latent viruses 14,15. However, there are exceptions. Some KS lesions transcribe almost the full complement of the viral transcriptome, and the KSHV-associated conditions plasmablastic variant of multicentric Castleman’s Disease (MCD) 16 and KSHV-associated Inflammatory Cytokine Syndrome (KICS)17,18 are associated with pronounced systemic viremia 18,19. KICS patients can have as many as five million KSHV genome copies per ml 20. KSHV viral load measurements in the blood may be utilized to detect MCD and KICS, which can co-occur in KS patients 17,21. While associated with concurrent primary effusion lymphoma (PEL), MCD, and KICS, systemic KSHV viral loads correlate with the number of KS lesions or other indicators of disease severity.

KSHV is shed with high frequency in the saliva of infected individuals, particularly while immune suppressed 22. Saliva is considered the most common route of transmission. Typically, the KSHV genome copy number in the saliva is considerably lower than that of EBV or human cytomegalovirus (HCMV), which also are transmitted by the oral route. This may explain the more limited transmission of KSHV in the general adult population compared to populations where saliva contact is more extensive, such as mother-to-child transmission and transmission among MSM 2325.

The multiple types of KS are summarized in Table 1. (i) Classic KS is a disease of older men around the Mediterranean basin and Saudi Arabia. Classic KS is independent of HIV infection and is concentrated in regions with a population prevalence of KSHV of 4–9% 26,27. Disease emergence of classic KS is age-related, perhaps a sign of immune senescence, although no immune biomarker has been identified that correlates with classic KS or can predict it. KSHV serology is not currently used as a screening tool for KS risk. Recent reports document cases of KS in younger HIV-negative gay men, often localized to the foot 2830, which would be consistent with the high KSHV transmission rate in this population. One could speculate that pre-exposure prophylaxis for HIV (Prep) without physical barriers does not protect against KSHV transmission.

Table 1:

Multiple Manifestations of Kaposi Sarcoma

Form age in years HIV Location
classic ≥60 Italy, Turkey, Saudi Arabia
endemic 0 – 16, ≥60 Africa, Xinjiang (China)
iatrogenic ≥60 global
AIDS (epidemic) 16 – 60 + US, Europe, Africa
pediatric-HIV 0 – 16 + Africa
endemic-HIV 0–60 + Africa
KS-IRIS 0–60 + global
Long-term cART ≥60 + global
Iatrogenic-HIV ≥60 + global
KICS 0 – 16 + global

(ii) Endemic KS is seen among the Kazakh and Uyghur ethnicities in Xinjiang (China) and in Eastern Africa (Kenia, Uganda, Malawi, Zambia, South Africa, and Tanzania). Pockets of high KSHV seroprevalence have also been reported in Amerindians 3133. In Eastern Africa, KS is the most common cancer in adult men and is also observed in children. Among the Uyghur, KSHV has also been associated with osteosarcoma, in addition to Kaposi Sarcoma 34. In endemic regions, the KSHV seroprevalence exceeds 50%, and seroconversion takes place before puberty, i.e., mother-to-child or intra-family transmission is the predominant route of infection. Endemic KS is seen in both HIV-positive and HIV-negative persons, old and young, men as well as women. Preliminary studies suggest that KSHV acquisition, if not KS disease, is associated with parasite co-infections in the region 35,36.

(iii) Iatrogenic KS is seen in solid organ transplant patients (HIV+ and HIV−), and KSHV may be transmitted by the transplanted organ itself 37. As expected, the prevalence of transplant KS tracks with both KSHV population seroprevalence and the number of kidney transplants in each country, with Italy and Saudi Arabia reporting the highest number of transplant KS cases.

(iv) AIDS-KS or epidemic KS was recognized as an AIDS-defining condition, next to Pneumocystis pneumonia, cryptococcus disease, and histoplasmosis 1. It is closely linked to HIV infection because of a shared pattern of transmission among high-risk individuals and because of HIV-induced immune deficiency. The HIV pandemic overlaps with different pre-existing classes of KS. Before the introduction of effective Prevention-of-Mother-to-Child-Transmission programs for HIV, pediatric HIV-KS rates were dramatically higher in KSHV endemic regions 38. Adult HIV-KS rates remain elevated in KSHV endemic regions since most PLWH worldwide reside in areas where KSHV was endemic before the introduction of HIV. In fact, treatment of HIV with cART sometimes leads to a flare-up of KS, a phenomenon termed Kaposi Sarcoma Immune Reconstitution Syndrome (KS-IRIS), which can be alleviated by concomitant oral etoposide 39.

Lastly, there are familial cases of KS. Here a person or family with inborn immune deficiency becomes exposed to KSHV and develops KS. KS has been reported in a person with Wescott-Aldrich syndrome as well as in persons carrying mutations in the STIM1, the gene encoding stromal interaction molecule 1, OX40, the co-stimulatory receptor expressed on activated T cells, MAGT1, STAT4, WAS, IFNGR1, and TNFRSF4 have been associated with KS4047. These are examples of genes where a single mutation can dramatically increase susceptibility to KSHV acquisition, KSHV reactivation, or KS disease. One would expect many more, less penetrant alleles to segregate in the population (reviewed in 48).

All types of KS are treated primarily with cytotoxic chemotherapy, mainly using a regimen developed for terminal AIDS-KS before cART was invented 49,50. In low- and middle-income countries (LMIC) where pegylated liposomal doxorubicin is not affordable in the public sector, free doxorubicin drug, vincristine, or paclitaxel are used 51. Newer experimental approaches are discussed below. There currently is no cure for KSHV infection latent and no vaccine against KS.

The most significant barrier to understanding KS and developing targeted KS therapies is the absence of small animal models for this tumor or even representative cell culture models. To this day, no one has successfully adapted KS tumor-derived cells to permanent growth in culture. This was not for lack of trying 5254. KS-derived explant cultures and KSHV-infected primary endothelial cells lose the viral genome in cell culture unless the viral episome is under strong selection, such as provided by a selectable marker or the particular genetic makeup of primary effusion lymphoma (PEL) 5559. PEL, unlike KS, has numerous genetic abnormalities, some of which, such as mutations in p53 60, were selected in response to chemotherapy, while others precede therapy. Most PEL also carries EBV 61. One could speculate that the PEL somatic mutations evolved in the presence of the latent KSHV genome, thus influencing KSHV maintenance, which is required for PEL survival.

From a clinical point of view, the disease manifestation of KS has not changed much since its initial discovery, and the treatment options have not advanced since the introduction of pegylated liposomal doxorubicin. One could argue, therefore, that KS is the same disease as it always was, only the circumstances under which the disease manifests have changed. AIDS-KS has become rarer in the US and Europe as cART has become readily available, and cancer diagnosis and therapy are accessible to most of the population. KSHV is firmly entrenched in vulnerable populations independent of HIV, and KS is now seen at a younger age. In Sub-Saharan Africa, where most PLWH and people living with KSHV reside, KS remains prevalent in the general population, and optimal therapy in the form of pegylated liposomal doxorubicin remain unavailable to most patients.

Histopathology of KS

KS is a tumor of dedifferentiated or transdifferentiated endothelial cells. This circumspect lineage description tries to coalesce several competing interpretations of the mature KS tumor cell’s nature and lineage of origin. The diagnosis of KS is based on the WHO definition of KS. The WHO pathology standard defines KS as “a locally aggressive endothelial proliferation that usually presents with cutaneous lesions in the form of multiple patches, plaques, or nodules, but it may also involve mucosal sites, lymph nodes, and visceral organs. KS is uniformly associated with HHV8 infection, and it represents an example of virus-induced vascular proliferation.” The WHO uses the presence of KSHV as the sole defining diagnostic criterium for KS.

Positivity for the KSHV genome or a KSHV protein encoded by the virus distinguishes KS from histologically similar, KSHV-negative lesions, such as angiosarcomas 62,63. The detection of KSHV DNA in KS lesions proved the association between this virus and this tumor beyond a reasonable doubt 64. It provided the basis for a new approach to KS diagnosis in low- and middle-income countries (LMIC) 65. The standard for establishing the presence of KSHV in a tumor lesion is immune histochemistry for the KSHV open-reading frame 73 protein, the so-called latency-associated nuclear antigen (LANA, LNA, or LANA-1) 6668. No other marker protein has been accepted for the clinical diagnosis of KS. No KS lesion has been described that would carry the viral genome but not express LANA. The LANA promoter is constitutively active in all cell types tested, and LANA is the only mRNA observed in every KSHV-infected cell 69,70.

Below we expand on two aspects of the WHO definition: the complexity of KSHV transcription in KS and the putative cell types associated with KSHV lesions. The genomic locus for LANA/ORF73 encompasses a series of genes: LANA, vCyc/ORF72, and vFLIP/ORF71. A single leftward transcript originating upstream of the LANA ORF covers the entire region. The vCyc protein is expressed by alternative splicing and from an internal promoter 71. The vFlip protein is expressed by an internal ribosome entry site on the vCyc-vFlip message 72. A common proximal 3’-poly adenylation site is used for the transcripts encoding these three proteins; however, a population of this transcript extends even further and terminates after the Kaposin/K12 gene. The Kaposin transcript represents the most common mRNA in KS tumors. The large intron of major latency transcript encodes the KSHV micro RNAs (miRNAs) 7375, except miR-K10a, encoded within the Kaposin ORF 76. The LANA transcript has been observed by in situ hybridization in KS tumors 77, and KSHV miRNAs have been found in KS tumors and PEL 78.

There exists considerable heterogeneity of viral transcription across and within KS lesions 15,7983. Every KS tumor transcribes the KSHV latency locus, and every KS tumor cell within a clinical lesion expresses the LANA protein. This correlation has biological plausibility since LANA is necessary and sufficient to maintain the KSHV plasmid within the nucleus of any infected cell 55,8486. Consistent with this ubiquitous expression pattern, the latency locus is free of repressive chromatin at all times 8790. On top of this minimal transcriptional state, which we term “tight latency” or latency I analogous to EBV, there exists a gradient of transcription patterns. These tight latency genes have been demonstrated in individual cells, in all KSHV-associated diseases, and all individual KS lesions.

However, additional genes are often transcribed in KS and KSHV-associated diseases as well. The data here are less complete and often limited by a lack of reagents. The antibody reagents to the LANA repeat region are more specific and sensitive than any other antibody against KSHV. The absence of evidence should not be taken as evidence of absence. In PEL, the LANA-2/vIRF3 locus is also consistently transcribed; the LANA-2/vIRF-3 protein is expressed - perhaps also vIRF1 and other genes in this locus. The region around the two lytic origins of replication also seems transcribed under an expanded set of conditions. This includes the viral IL-6 protein, which is typically for MCD 91,92. Recent single-cell and tumor profiling data suggest the existence of additional actively transcribed regions on the genome in KS and PEL 14,15,70, a semi-stable state that we describe as “extended latency” or latency II. These include, for instance, the nut-1 nuclear transcript 77. In addition, the K15 transcript is detectable in PEL, and the K15 protein has been detected in many KS lesions93. K15 is located on the right-hand side of the genome. It is variable and differentially spliced, making the RNA difficult to detect 94,95.

Lastly, about one-third of KS lesions harbor transcripts across the entire viral genome 14. These prototypical patterns are observed in any set of KS biopsies and any collection of KSHV-infected cells with some variations. The clinical significance of these different transcription patterns is unknown. Still, differential viral transcription and the different cellular compositions of individual KS lesions may explain the clinical observation that all skin KS lesions in patients on systemic therapy and cART, today, respond equally.

Another feature of KSHV is worth mentioning in this context. All KSHV miRNAs have been observed in circulating extracellular vesicles or exosomes isolated from tissue culture supernatant, from KS patients, in the context of viral infection, and from cells that ectopically expressed only the KSHV miRNAs 96. Detecting the viral miRNAs in EVs or the linear, encapsulated viral genome in plasma thus constitutes a biomarker for KSHV infection. Because EV-encased miRNAs can travel independently of the virus and are not restricted by virus-specific entry receptors, they can induce “KS-like” phenotypes in uninfected endothelial cells 97,98. This series of experiments independently confirms that much of the lineage reprogramming induced by KSHV can be attributed to the KSHV miRNAs, most likely in conjunction with paracrine host factors, such as PDGF 98,99. This phenotype thus increases the heterogeneity of the KS lesion even further. Every KS lesion is an unstable mixture of cells infected with KSHV, not infected with KSHV but reprogramed by EV-transduced KSHV miRNAs, or neither infected with the virus nor carrying the viral miRNAs but exposed to soluble paracrine factors, notably vIL6, IL6, VEGF, and PDGF. In sum, KS heterogeneity is indistinguishable by morphology 63,100102.

Pathologists distinguish patch, plaque, and nodular lesions chiefly based on the degree of spindle cell nests in a section and neoangiogenesis 103,104. Confusingly, the same terms are also general dermatology terms and have been used to describe the gross appearance of KS skin lesions. In other contexts, the terms flat, raised, and nodular are used informally to describe KS skin lesions 105. In response to systemic cytotoxic chemotherapy, the standard of care in KS, lesions tend to flatten before they disappear. As noted above, not all skin lesions on a patient respond equally to standard therapy, and not all patients respond. This inconsistent behavior may be due to the cellular and molecular heterogeneity among KS lesions in the same patient. Complete responses are rare; partial responses and stable disease represent the most commonly observed outcome, particularly in KSHV-endemic regions 51,106,107. At times these response classifications are subject to observer bias. Detailed studies of how the overt clinical appearance corresponds to histopathology are missing. The molecular and clinical heterogeneity of KS lesions has foiled clinical trials for multiple targeted agents. Multiple agents such as the VEGF-1 inhibitor Bevacizumab, the mTOR inhibitor sirolimus/rapamycin 108,109 and receptor-tyrosine kinase inhibitors, such as Imatinib and sorafenib 110112 sometimes show significant efficacy as single agents in small trials and some patients. Still, it is unclear why the other KS patients do not respond. Our lack of a precise understanding of the molecular, histopathological, immune components, and clinical heterogeneity of KS severely hampers the development of rational and targeted therapies.

The lineage of the KS tumor cell

Although the WHO has classified KS as an “endothelial proliferation,” it has not endorsed any particular histochemical lineage markers for KS diagnosis. Sarcoma biomarkers and endothelial and mesenchymal lineage markers have all been observed in KS lesions. This led to considerable confusion in the field but has not yielded clinically actionable insights 9. Experimental studies with the KSHV virus in culture added to this mélange of observations. It is essential to keep these two lines of evidence, observations in clinical lesions and experimental infections, separate.

Hong et al. 113 and Wang et al. 114 reported the first host transcriptional profile of KS lesions. These studies and others 102 support the notion that KS is a tumor of endothelial cell lineage, but not just composed of purely endothelial cells. This sentiment is confirmed by immune histochemistry. KS lesions stain positive for lymphatic endothelial cell lineage markers, such as lymphatic vessel endothelial receptor 1 (Lyve-1), VEGFR3, and Podoplanin, van Willebrand factor, CD31/ PECAM-1, CD34. Prox1 and VEGFR3 are perhaps the most specific markers, as they are typically expressed only on lymphatic and not blood endothelial cells 115. In KS lesions, LANA-positive cells express VEGFR-3 (as do some LANA-negative cells) 116. Because KS is highly vascularized, and most lesions undergo active angiogenesis and vessel remodeling, it is difficult to decide for each cell whether it originated from the nest of KS tumor cells (and perhaps lost the virus as it differentiated) or migrated into the lesions as a CD34+/CD31+ circulating endothelial cell precursor cells and then became KSHV-infected and reprogrammed within the KS microenvironment 117. In KS, unlike any other tumor, there is no distinction between the tumor neovasculature and the tumor itself.

Differentiated endothelial cell markers are not the only ones expressed in KS lesions. Gill and colleagues 118 noted that the Notch isotype expression in KS does not match “normal,” i.e., fully differentiated endothelial cells. In culture, KSHV can induce Notch and endothelial-to-mesenchymal transition 119,120. This observation prompted research into inhibiting IC-Notch signaling in KSHV diseases. Unfortunately, the most clinically developed candidate had an unacceptable safety profile. Other markers, such as PDGF-1, have been described in KS or KSHV-infected cells typically associated with mesenchymal lineage or mesenchymal stem cells 99,121,122. This is expected as “nests” of KSHV-infected tumor cells are embedded in a matrix of stromal cells. Depending on the stage of the particular lesion - patch, plaque, nodular-stromal cells, or even immune cells may make up most of the lesion.

Infection of purified cell populations with KSHV in culture has generated evidence supporting multiple scenarios. KSHV can infect and persist in multiple cell types, perhaps more efficiently in stem cells but also HEK293 cells 117,123,124. In pure culture, infection with KSHV can differentiate precursor cells into lymphatic endothelial cells and transdifferentiate blood endothelial cells (HUVEC) into lymphatic endothelial cells and vice versa 113,114,125. The endothelial lineage markers PROX1 and SOX18 have roles in the KSHV life cycle 126. It was also reported that KSHV could differentiate primitive mesenchymal cells into endothelial cells 127. These culture experiments reinforce the “chicken and egg” problem regarding the trans*differentiation of precursors into more differentiated cells or vice versa. Does KSHV infect mesenchymal stem cells and induce the extraordinary expression of some endothelial markers, or did KSHV infect mature endothelial cells and dedifferentiate them into more primitive precursors, common ancestors of the mesenchymal/endothelial cell lineage? These in-culture infection experiments suggest that both directions are possible, and that the virus is apt at generating the perfect cellular environment for persistence.

Individual KSHV viral genes, such as K1, vIL6, and vGPCR, in isolation, can also dedifferentiate, transdifferentiate and, in rodent systems, fully transform mature endothelial cells 128130. The KSHV miRNAs in isolation induce trans-differentiation and hallmarks of transformation 76,78,97,98,131,132. In culture, KSHV can transform both endothelial and mesenchymal lineage precursors, as well as experimental cell models, to the point that they form tumors as xenografts in immune-deficient mice 102,124,133136. These experiments support the classification of KSHV as a bona fide human tumor virus; however, tissue culture systems, by design, are finely tuned to respond to pro-growth signals with drastic changes in cell phenotypes. Some, such as NIH3T3 cells, were evolved to score a single hit (mtP53) or dual hit (Myc and mtRas) oncogenes. In culture, both overexpression and mutational activation of single proteins induce transformation. This has been demonstrated for Myc, Ras/h-Ras, GPCR/vGPCR, huIL6/vIL6, and members of the PI3k/mTOR pathway 137. Myc is the prime example as either amplification, translocation, stabilization, or mutation induces similar phenotypes in susceptible experimental systems and is found in tumors 58,138,139. The question is not which pathway is more important but which constellation of pathways is active in a particular KS lesion and how it can be targeted therapeutically.

KSHV viral load as a diagnostic marker.

Oncogenic viruses cause about one fifth of all cancers worldwide. These viruses are present in all tumor cells, and viral genes (proteins and miRNAs) drive the hallmarks of cancer 140. One would think, therefore, that viral load measurements constitute a biomarker for viral cancers. This is indeed the case for human papillomavirus (HPV)-associated cervical cancer, where the persistence of viral DNA copies of the high-risk types has become the primary screening tool for preventing the disease. Detecting high-risk HPV is a clinically proven predictive biomarker for cervical cancer and a prognostic marker in HPV-positive oropharyngeal cancer 141. Likewise, viral load is an established measure for most acute viral diseases. In HIV-infected patients, viral genome copy number is the sole determinant of clinical intervention, i.e., cART initiation. Increases in circulating HCMV are the clinically actionable measure to start and stop antiviral therapy in transplant patients. In HCMV, viral load is measured either by genome copy number measured by QPCR or leucocyte antigenemia measured by pp65 levels142. Viral load is routinely measured in nasal swabs for respiratory viruses. The COVID-19 pandemic has broadly demonstrated the utility of virus detection in directing public health interventions and initiating antiviral therapy. In sum, a solid body of precedence exists for the utility of viral loads as predictive and prognostic biomarkers in any viral disease.

The situation regarding KS is more complicated (reviewed in 5). Every KS tumor cell carries one or more copies of the KSHV genome and expresses some viral proteins and all viral miRNAs. The presence of KSHV DNA in KS biopsies is under consideration for a rapid field test in KS endemic areas 65. Likewise, detecting the EBV EBER small non-coding RNAs constitutes the gold standard for diagnosing EBV lymphoma 143. Yet, herpesvirus loads in plasma or saliva do not have broadly prognostic or predictive power. The problem with using KSHV viral load measurements in blood as biomarkers for cancer is that even cancer-free people have circulating virus in their blood and saliva. Genome positivity for any herpesvirus in blood or saliva is a measure of infection, not an imminent disease; rather, herpesviruses are shed asymptomatically for life.

There have been many attempts to demonstrate a correlation between KSHV systemic copy number and KS 144. Reviewing these efforts highlight both difficulties and opportunities. There are three sources of viral DNA in circulation: (i) inside healthy, circulating latently infected cells, (ii) inside infectious virions and non-infectious, sometimes called interfering, particles, and (iii) as part of naked circulating tumor DNA released by the lysis of tumor cells or reactivating latently infected normal cells. These need to be distinguished when testing for clinical associations.

Healthy-latently infected cells characterize the KSHV latent reservoir in normal individuals 145. We have studied the utility of the KSHV miRNAs as a marker for latent infection and tumor load 96. Unlike viral DNA, the KSHV miRNA levels do not change markedly during viral reactivation, except for miRNA K12–11. The viral miRNAs are constantly transcribed and processed from the KSHV latency transcript. The mature, correctly processed viral miRNAs are excreted into circulation. Therefore, the viral miRNAs are protected from RNAses. KSHV miRNA levels in blood correlated with tumor burden and the number of healthy, latently infected cells. As of yet, neither approach has entered clinical practice. Developing KSHV viral load measurements in blood represent a diagnostic opportunity for rapid testing and monitoring of therapy response that has not been realized.

Virus-Immune Interactions

Innate immunity is the first line of defense against invading pathogens. Pathogen-associated molecular patterns (PAMPs) on incoming microbes are detected by pathogen recognition receptors (PRRs). Activation of PRRs triggers signal transduction pathways, resulting in the production of interferon and inflammatory cytokines 146. The KSHV genome encodes many immunomodulatory proteins that prevent antiviral responses from PRRs 147,148. This immunomodulation allows viral persistence in the host 149 and is reviewed here.

First, toll-like receptors are PRRs that detect incoming microbes in endosomes (TLR3/7/8/9) or on the plasma membrane (TLR1/2/4/5/6/10) 150. Infection with KSHV activates TLR3 151, but TLR3-mediated interferon induction is diminished by KSHV viral interferon regulatory factors (vIRFs) 152. The KSHV lytic switch protein, RTA, promotes the degradation of the TLR3 adaptor protein, TRIF 153, and also suppresses gene expression of Myeloid differentiation primary response 88 (MyD88), TLR4 adaptor protein 154.

Second, the nucleotide-binding oligomerization domain-like receptors (NOD-like receptors, or NLRs) are present in the cytoplasm and can also recognize PAMPs. Some NLRs are components of inflammasomes, which upon activation, induce the production of inflammatory cytokines like IL-1 or IL-18 150. While the early lytic protein, KSHV ORF45, activates the NLRP1 inflammasome, the tegument protein, KSHV ORF63, can inhibit NLR proteins 155,156. Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) have traditionally been considered RNA sensors but can also detect DNA viruses like KSHV 150,157160. RIG-I is suppressed by the KSHV deubiquitinase ORF64, which prevents RIG-I ubiquitination and activation 161.

Third, KSHV inhibits cytosolic DNA sensing pathways, including the cGAS-STING pathway. STING on the ER membrane is bound and activated by the cyclic GMP-AMP (cGAMP) generated by activated cGAS. KSHV ORF52 binds to cGAS and prevents the synthesis of cGAMP 162. Additionally, LANA binds cGAS, preventing TBK1 from being activated 163, and vIRF1 binds to STING and prevents TBK1 recruitment 164. In lay terms, an intricate battle ensues with each infection event between the virus and the cell’s innate host response. KS is an outcome where the virus wins this battle. We do not know if that represents the majority or a small minority of infection events. We do not know which other factors are involved, but some interchanges are governed by inborn defects and polymorphisms in the human population 41,42,45.

KSHV modulates cell-mediated immunity. Natural killer (NK) cells, T cells, dendritic cells, and macrophages scour the host for pathogens. Many of these immune cells are capable of identifying and killing virus-infected cells. However, KSHV encodes countermeasures. Many KSHV proteins inhibit NK- or T- cell-mediated killing. For example, the viral ubiquitin ligase KSHV K3 promotes the degradation of MHC-I, CD1d, CD31, and IFN-R1, which prevents CD8+ T lymphocytes from becoming activated and recognizing infected cells. Another viral ubiquitin ligase, KSHV K5, also induces degradation of MHC-I, CD54, B7–2, CD1d, MICA, and MICB, thereby impairing CD8+ T cell and NK cell activity 165. In addition, MICB expression is downregulated by the KSHV microRNA miR-K12–7 166. KSHV also subverts CD4+ T cell-mediated immunity. KSHV RTA upregulates an MHC-II antagonist, i.e., membrane-associated RING-CH (MARCH8), while promoting the proteasomal degradation of MHC-II 167. Autophagy is necessary for MHC-II antigen display, but viral B-cell leukemia/lymphoma 2 (vBcl2) prevents it 168. In addition, IFN and class II MHC transactivator (CIITA) is downregulated by vIRF3, which reduces MHC-II presentation 169. KSHV LANA binds to the CIITA promoter and prevents IRF4 from activating it, which reduces MHC-II expression 170.

The cell-extrinsic effects of KSHV on the immune system are, in part, mediated by secreted viral proteins. KSHV encodes three cellular chemokine viral homologs, vCCL1 to 3 (also known as vMIP-I to III) 149. vCCL3 preferentially stimulates chemotaxis in Th2 cells compared to Th1 cells, indicating that viral chemokines may contribute to developing the KS microenvironment’s distinctively Th2-skewed milieu 171. vCCL2 binds to CX3C chemokine receptor 1 (CX3CR1) and CCR5, suppressing the natural ligands of these receptors and preventing the migration of NK cells 172. vCCL2 also induces CCR3 and CCR8 expression in order to attract Th2 cells to KS lesions, while inhibiting CCR1 and CCR5 activation 171. In addition, KSHV encodes an IL-6 viral homolog (vIL-6). vIL-6 signals through gp130 dimers, whereas IL-6 requires IL-6R and gp130 for signaling 173. Both cytokines activate Janus kinase (JAK)-STAT signaling, primarily through STAT3, which is linked to angiogenesis, migration, differentiation, and cell proliferation 174,175. The vIL-6 promotes increased IL-6 release, which increases B cell proliferation and causes flare-ups of MCD or KS/PEL 176. The survival and proliferation of PEL cells are aided by low levels of latent vIL-6 expression 177. It has been demonstrated that vIL-6 increases the enzyme activation-induced cytidine deaminase (AID) production in activated B cells, which increases the rate of class-switch recombination 178.

Immune targeting therapies for KS and KSHV-associated diseases

Some label the 21st century as the century when cancer research learned how to appropriate the immune system for cancer therapy. KS and KSHV-associated cancers represent perfect targets, as the viral proteins are considered non-self, and KS and KSHV heavily depend on the tumor and immune microenvironment. After all, transplant KS resolves in response to a reduction in immune suppressive dose. A significant fraction of limited KS resolves after HIV-associated immune deficiency is overcome by cART (eventually, the cancer returns in both scenarios).

Anti-IL-6 antibodies were first tested as a therapy for idiopathic MCD, which is known to express significant amounts of human IL-6. Siltuximab, an anti-IL-6 antibody, was effective against KSHV-associated MCD 179. Tocilizumab, an anti-IL-6R antibody, was tested in a small cohort of KSHV-associated MCD patients and was found to show activity and be safe 180.

Programmed death ligand 1 (PD-L1) is an inhibitory molecule overexpressed on many different tumor types. PD-L1 binds to its receptor, programmed cell death protein 1 (PD1), to suppress immune responses. Increased PD-L1 expression in monocytes induced by KSHV infection may help KSHV evade the immune system 181. PD1 is persistently increased in NK cells recovered from KS patients, indicating an exhausted phenotype 182. Nivolumab and pembrolizumab are examples of anti-PD1 antibodies that reduce tumor size in KS patients harboring HIV 183185 as well as in classic and endemic KS 186. Pembrolizumab was previously shown to have an acceptable safety profile in HIV patients with cancer 185.

Further studies are ongoing, as it is unclear how these drugs affect HIV latency 187,188. These include trials investigating intralesional injections of Nivolumab in cutaneous KS (NCT03316274) and pembrolizumab in combination with anti-retroviral therapy in HIV-infected individuals with cancer (NCT02595866). Another inhibitory modulator of T cells is cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) 189. Ipilimumab is an anti-CTLA-4 antibody and, in combination with Nivolumab in KS patients, displayed good efficacy 190,191.

Pomalidomide and Lenalidomide 13,192,193, both derivatives of Thalidomide, represent the only new drug class approved for treating KS in the last 20 years. Pomalidomide was safe and active in KS patients with and without HIV, with an overall response rate of seventy-one percent 194. This led to accelerated approval in the USA. Several more extensive studies are ongoing worldwide (NCT04577755, NCT02659930, NCT03601806). These will establish the efficacy of pomalidomide across diverse populations of KS patients. Their primary target is Cereblon, which is an essential gene in PEL 195. In addition to its cell intrinsic role in survival, pomalidomide modulates the immune response at both a cellular and systemic level 196. Pomalidomide restores B7–2, ICAM-1, and MHC1 levels in latent and lytic PEL cells 197. In addition, this drug also prevents tumor cells from upregulating PD-L1 198. It may kill virally infected cells in the KS lesion directly, render them visible to the adaptive immune system, or modulate the inflammatory lesion microenvironment to the point where it can no longer sustain tumor cell proliferation by intrinsic or paracrine modulators.

In sum, we hope that this review has highlighted that most patients that develop KS today are nothing like the AIDS-KS patients that led to the discovery of KSHV and the formulation of chemotherapy-based KS treatments. KS patients today are diverse; they experience KS under different circumstances and exhibit different but still underdefined phenotypes of KSHV infection. We lack a clear understanding of what exactly goes on in the KS lesion microenvironment and which are the rate-limiting molecular processes that maintain this cancer. We have no outcome markers beyond lesion measurements and no predictive or prognostic molecular tests. The KS and HIV-KS epidemic cannot be considered solved; it remains rampant, particularly in LMIC environments where the best standard of chemotherapy care is not widely affordable. We are still learning to optimally target KSHV to treat KS and monitor KSHV to predict KS. Pomalidomide and cART provide a new oral treatment modality for KS.

Acknowledgements

This work was supported by public health service grants CA019014, CA23958 to BD and DPD. Neither the funders nor the University of North Carolina had any role in study design, data collection and interpretation, or the opinions represented here. We than Anthony B. Eason and Razia Moorad for critical reading.

Footnotes

Conflict of Interest

The authors declare no conflicts.

Data availability statement

As this is a review, the data are available in PubMed and as listed in references.

References

  • 1.Centers for Disease C Kaposi’s sarcoma and Pneumocystis pneumonia among homosexual men--New York City and California. MMWR Morb Mortal Wkly Rep. 1981;30(25):305–308. [PubMed] [Google Scholar]
  • 2.Maurer T, Ponte M, Leslie K. HIV-associated Kaposi’s sarcoma with a high CD4 count and a low viral load. N Engl J Med. 2007;357(13):1352–1353. [DOI] [PubMed] [Google Scholar]
  • 3.Krown SE, Lee JY, Dittmer DP, Consortium AM. More on HIV-associated Kaposi’s sarcoma. N Engl J Med. 2008;358(5):535–536; author reply 536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–249. [DOI] [PubMed] [Google Scholar]
  • 5.Cesarman E, Damania B, Krown SE, Martin J, Bower M, Whitby D. Kaposi sarcoma. Nat Rev Dis Primers. 2019;5(1):9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lothe F Multiple idiopathic haemorrhagic sarcoma of Kaposi in Uganda. Acta Unio Int Contra Cancrum. 1960;16:1447–1451. [PubMed] [Google Scholar]
  • 7.Chang Y, Cesarman E, Pessin MS, et al. Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi’s sarcoma. Science. 1994;266(5192):1865–1869. [DOI] [PubMed] [Google Scholar]
  • 8.Luo Q, Pfeiffer RM, Noone AM, Horner MJ, Engels EA, Shiels MS. Years of life lost to cancer among the US HIV population, 2006–2015. AIDS. 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Reid E, Suneja G, Ambinder RF, et al. AIDS-Related Kaposi Sarcoma, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2019;17(2):171–189. [DOI] [PubMed] [Google Scholar]
  • 10.El-Mallawany NK, Villiera J, Kamiyango W, et al. Endemic Kaposi sarcoma in HIV-negative children and adolescents: an evaluation of overlapping and distinct clinical features in comparison with HIV-related disease. Infect Agent Cancer. 2018;13:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cao Y, Minhas V, Tan X, et al. High prevalence of early childhood infection by Kaposi’s sarcoma-associated herpesvirus in a minority population in China. Clin Microbiol Infect. 2014;20(5):475–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.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(4):233–241. [DOI] [PubMed] [Google Scholar]
  • 13.Reid EG, Shimabukuro KA, Moore P, et al. AMC-070: Lenalidomide is Safe and Effective in HIV-associated Kaposi Sarcoma. Clin Cancer Res. 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hosseinipour MC, Sweet KM, Xiong J, et al. Viral profiling identifies multiple subtypes of Kaposi’s sarcoma. MBio. 2014;5(5):e01633–01614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Dittmer DP. Restricted Kaposi’s sarcoma (KS) herpesvirus transcription in KS lesions from patients on successful antiretroviral therapy. MBio. 2011;2(6):e00138–00111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Cesarman E, Chang Y, Moore PS, Said JW, Knowles DM. Kaposi’s sarcoma-associated herpesvirus-like DNA sequences in AIDS-related body-cavity-based lymphomas. N Engl J Med. 1995;332(18):1186–1191. [DOI] [PubMed] [Google Scholar]
  • 17.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(6):730–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.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(3):350–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.El-Mallawany NK, Kamiyango W, Villiera J, et al. Kaposi Sarcoma Herpesvirus Inflammatory Cytokine Syndrome-like Clinical Presentation in Human Immunodeficiency Virus-infected Children in Malawi. Clin Infect Dis. 2019;69(11):2022–2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Tamburro KM, Yang D, Poisson J, et al. Vironome of Kaposi sarcoma associated herpesvirus-inflammatory cytokine syndrome in an AIDS patient reveals co-infection of human herpesvirus 8 and human herpesvirus 6A. Virology. 2012;433(1):220–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ramaswami R, Lurain K, Polizzotto MN, et al. Characteristics and outcomes of KSHV-associated multicentric Castleman disease with or without other KSHV diseases. Blood Adv. 2021;5(6):1660–1670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Koelle DM, Huang ML, Chandran B, Vieira J, Piepkorn M, Corey L. Frequent detection of Kaposi’s sarcoma-associated herpesvirus (human herpesvirus 8) DNA in saliva of human immunodeficiency virus-infected men: clinical and immunologic correlates. J Infect Dis. 1997;176(1):94–102. [DOI] [PubMed] [Google Scholar]
  • 23.Martin JN, Ganem DE, Osmond DH, Page-Shafer KA, Macrae D, Kedes DH. Sexual transmission and the natural history of human herpesvirus 8 infection. N Engl J Med. 1998;338(14):948–954. [DOI] [PubMed] [Google Scholar]
  • 24.Newton R, Labo N, Wakeham K, et al. Determinants of Gammaherpesvirus Shedding in Saliva Among Ugandan Children and Their Mothers. J Infect Dis. 2018;218(6):892–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Malope BI, Pfeiffer RM, Mbisa G, et al. Transmission of Kaposi sarcoma-associated herpesvirus between mothers and children in a South African population. J Acquir Immune Defic Syndr. 2007;44(3):351–355. [DOI] [PubMed] [Google Scholar]
  • 26.Whitby D, Luppi M, Barozzi P, Boshoff C, Weiss RA, Torelli G. Human herpesvirus 8 seroprevalence in blood donors and lymphoma patients from different regions of Italy. J Natl Cancer Inst. 1998;90(5):395–397. [DOI] [PubMed] [Google Scholar]
  • 27.Whitby D, Luppi M, Sabin C, et al. Detection of antibodies to human herpesvirus 8 in Italian children: evidence for horizontal transmission. Br J Cancer. 2000;82(3):702–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Casper C, Carrell D, Miller KG, et al. HIV serodiscordant sex partners and the prevalence of human herpesvirus 8 infection among HIV negative men who have sex with men: baseline data from the EXPLORE Study. Sex Transm Infect. 2006;82(3):229–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lanternier F, Lebbe C, Schartz N, et al. Kaposi’s sarcoma in HIV-negative men having sex with men. AIDS. 2008;22(10):1163–1168. [DOI] [PubMed] [Google Scholar]
  • 30.Swali R, Limmer A, Tyring SK. Kaposi Sarcoma of the Medial Foot in an MSM, HIV-Negative Man: A Fifth Clinical Variant. J Clin Aesthet Dermatol. 2020;13(10):42–44. [PMC free article] [PubMed] [Google Scholar]
  • 31.Biggar RJ, Whitby D, Marshall V, Linhares AC, Black F. Human herpesvirus 8 in Brazilian Amerindians: a hyperendemic population with a new subtype. J Infect Dis. 2000;181(5):1562–1568. [DOI] [PubMed] [Google Scholar]
  • 32.Whitby D, Marshall VA, Bagni RK, et al. Genotypic characterization of Kaposi’s sarcoma-associated herpesvirus in asymptomatic infected subjects from isolated populations. J Gen Virol. 2004;85(Pt 1):155–163. [DOI] [PubMed] [Google Scholar]
  • 33.Lopes AO, Spitz N, Reis CRS, de Paula VS. Update of the global distribution of human gammaherpesvirus 8 genotypes. Sci Rep. 2021;11(1):7640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chen Q, Chen J, Li Y, et al. Kaposi’s sarcoma herpesvirus is associated with osteosarcoma in Xinjiang populations. Proc Natl Acad Sci U S A. 2021;118(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Oluoch PO, Oduor CI, Forconi CS, et al. Kaposi Sarcoma-Associated Herpesvirus Infection and Endemic Burkitt Lymphoma. J Infect Dis. 2020;222(1):111–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Sabourin KR, Daud I, Ogolla S, et al. Malaria Is Associated With Kaposi Sarcoma-Associated Herpesvirus Seroconversion in a Cohort of Western Kenyan Children. J Infect Dis. 2021;224(2):303–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Barozzi P, Luppi M, Facchetti F, et al. Post-transplant Kaposi sarcoma originates from the seeding of donor-derived progenitors. Nat Med. 2003;9(5):554–561. [DOI] [PubMed] [Google Scholar]
  • 38.El-Mallawany NK, Mehta PS, Kamiyango W, et al. KSHV viral load and Interleukin-6 in HIV-associated pediatric Kaposi sarcoma-Exploring the role of lytic activation in driving the unique clinical features seen in endemic regions. Int J Cancer. 2019;144(1):110–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Nyirenda M, Ngongondo M, Kang M, et al. Early Progression and Immune Reconstitution Inflammatory Syndrome During Treatment of Mild-To-Moderate Kaposi Sarcoma in Sub-Saharan Africa and South America: Incidence, Long-Term Outcomes, and Effects of Early Chemotherapy. J Acquir Immune Defic Syndr. 2020;84(4):422–429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Jackson CC, Dickson MA, Sadjadi M, et al. Kaposi Sarcoma of Childhood: Inborn or Acquired Immunodeficiency to Oncogenic HHV-8. Pediatric blood & cancer. 2016;63(3):392–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Aavikko M, Kaasinen E, Nieminen JK, et al. Whole-Genome Sequencing Identifies STAT4 as a Putative Susceptibility Gene in Classic Kaposi Sarcoma. J Infect Dis. 2015;211(11):1842–1851. [DOI] [PubMed] [Google Scholar]
  • 42.Byun M, Ma CS, Akcay A, et al. Inherited human OX40 deficiency underlying classic Kaposi sarcoma of childhood. J Exp Med. 2013;210(9):1743–1759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Leroy S, Moshous D, Cassar O, et al. Multicentric Castleman disease in an HHV8-infected child born to consanguineous parents with systematic review. Pediatrics. 2012;129(1):e199–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Sahin G, Palanduz A, Aydogan G, et al. Classic Kaposi sarcoma in 3 unrelated Turkish children born to consanguineous kindreds. Pediatrics. 2010;125(3):e704–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Byun M, Abhyankar A, Lelarge V, et al. Whole-exome sequencing-based discovery of STIM1 deficiency in a child with fatal classic Kaposi sarcoma. J Exp Med. 2010;207(11):2307–2312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Picard C, Mellouli F, Duprez R, et al. Kaposi’s sarcoma in a child with Wiskott-Aldrich syndrome. Eur J Pediatr. 2006;165(7):453–457. [DOI] [PubMed] [Google Scholar]
  • 47.Brigida I, Chiriaco M, Di Cesare S, et al. Large Deletion of MAGT1 Gene in a Patient with Classic Kaposi Sarcoma, CD4 Lymphopenia, and EBV Infection. J Clin Immunol. 2017;37(1):32–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Blumenthal MJ, Cornejo Castro EM, Whitby D, Katz AA, Schafer G. Evidence for altered host genetic factors in KSHV infection and KSHV-related disease development. Rev Med Virol. 2021;31(2):e2160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Stewart S, Jablonowski H, Goebel FD, et al. Randomized comparative trial of pegylated liposomal doxorubicin versus bleomycin and vincristine in the treatment of AIDS-related Kaposi’s sarcoma. International Pegylated Liposomal Doxorubicin Study Group. J Clin Oncol. 1998;16(2):683–691. [DOI] [PubMed] [Google Scholar]
  • 50.Northfelt DW, Dezube BJ, Thommes JA, et al. Pegylated-liposomal doxorubicin versus doxorubicin, bleomycin, and vincristine in the treatment of AIDS-related Kaposi’s sarcoma: Results of a randomized phase III clinical trial. Journal of Clinical Oncology. 1998;16(7):2445–2451. [DOI] [PubMed] [Google Scholar]
  • 51.Krown SE, Moser CB, MacPhail P, et al. Treatment of advanced AIDS-associated Kaposi sarcoma in resource-limited settings: a three-arm, open-label, randomised, non-inferiority trial. Lancet. 2020;395(10231):1195–1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Delli Bovi P, Donti E, Knowles DM 2nd, et al. Presence of chromosomal abnormalities and lack of AIDS retrovirus DNA sequences in AIDS-associated Kaposi’s sarcoma. Cancer Res. 1986;46(12 Pt 1):6333–6338. [PubMed] [Google Scholar]
  • 53.An FQ, Folarin HM, Compitello N, et al. Long-term-infected telomerase-immortalized endothelial cells: a model for Kaposi’s sarcoma-associated herpesvirus latency in vitro and in vivo. J Virol. 2006;80(10):4833–4846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Grundhoff A, Ganem D. Inefficient establishment of KSHV latency suggests an additional role for continued lytic replication in Kaposi sarcoma pathogenesis. J Clin Invest. 2004;113(1):124–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Bigi R, Landis JT, An H, Caro-Vegas C, Raab-Traub N, Dittmer DP. Epstein-Barr virus enhances genome maintenance of Kaposi sarcoma-associated herpesvirus. Proc Natl Acad Sci U S A. 2018;115(48):E11379–E11387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Roy D, Sin SH, Damania B, Dittmer DP. Tumor suppressor genes FHIT and WWOX are deleted in primary effusion lymphoma (PEL) cell lines. Blood. 2011;118(7):e32–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yang D, Chen W, Xiong J, Sherrod CJ, Henry DH, Dittmer DP . Interleukin 1 receptor-associated kinase 1 (IRAK1) mutation is a common, essential driver for Kaposi sarcoma herpesvirus lymphoma. Proc Natl Acad Sci U S A. 2014;111(44):E4762–4768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bubman D, Guasparri I, Cesarman E. Deregulation of c-Myc in primary effusion lymphoma by Kaposi’s sarcoma herpesvirus latency-associated nuclear antigen. Oncogene. 2007;26(34):4979–4986. [DOI] [PubMed] [Google Scholar]
  • 59.Luan SL, Boulanger E, Ye H, et al. Primary effusion lymphoma: genomic profiling revealed amplification of SELPLG and CORO1C encoding for proteins important for cell migration. J Pathol. 2010;222(2):166–179. [DOI] [PubMed] [Google Scholar]
  • 60.Petre CE, Sin SH, Dittmer DP. Functional p53 signaling in Kaposi’s sarcoma-associated herpesvirus lymphomas: implications for therapy. J Virol. 2007;81(4):1912–1922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Faure A, Hayes M, Sugden B. How Kaposi’s sarcoma-associated herpesvirus stably transforms peripheral B cells towards lymphomagenesis. Proc Natl Acad Sci U S A. 2019;116(33):16519–16528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Young RJ, Brown NJ, Reed MW, Hughes D, Woll PJ. Angiosarcoma. Lancet Oncol. 2010;11(10):983–991. [DOI] [PubMed] [Google Scholar]
  • 63.Khan JA, Maki RG, Ravi V. Pathologic Angiogenesis of Malignant Vascular Sarcomas: Implications for Treatment. J Clin Oncol. 2018;36(2):194–201. [DOI] [PubMed] [Google Scholar]
  • 64.Moore PS, Chang Y. Detection of herpesvirus-like DNA sequences in Kaposi’s sarcoma in patients with and those without HIV infection. N Engl J Med. 1995;332(18):1181–1185. [DOI] [PubMed] [Google Scholar]
  • 65.Snodgrass R, Gardner A, Jiang L, Fu C, Cesarman E, Erickson D. KS-Detect - Validation of Solar Thermal PCR for the Diagnosis of Kaposi’s Sarcoma Using Pseudo-Biopsy Samples. PLoS One. 2016;11(1):e0147636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kedes DH, Lagunoff M, Renne R, Ganem D. Identification of the gene encoding the major latency-associated nuclear antigen of the Kaposi’s sarcoma-associated herpesvirus. J Clin Invest. 1997;100(10):2606–2610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kellam P, Boshoff C, Whitby D, Matthews S, Weiss RA, Talbot SJ. Identification of a major latent nuclear antigen, LNA-1, in the human herpesvirus 8 genome. J Hum Virol. 1997;1(1):19–29. [PubMed] [Google Scholar]
  • 68.Rainbow L, Platt GM, Simpson GR, et al. The 222- to 234-kilodalton latent nuclear protein (LNA) of Kaposi’s sarcoma-associated herpesvirus (human herpesvirus 8) is encoded by orf73 and is a component of the latency-associated nuclear antigen. J Virol. 1997;71(8):5915–5921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Dittmer D, Lagunoff M, Renne R, Staskus K, Haase A, Ganem D. A cluster of latently expressed genes in Kaposi’s sarcoma-associated herpesvirus. J Virol. 1998;72(10):8309–8315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Landis JT, Tuck R, Pan Y, et al. Evidence for Multiple Subpopulations of Herpesvirus-Latently Infected Cells. mBio. 2022:e0347321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Pearce M, Matsumura S, Wilson AC. Transcripts encoding K12, v-FLIP, v-cyclin, and the microRNA cluster of Kaposi’s sarcoma-associated herpesvirus originate from a common promoter. J Virol. 2005;79(22):14457–14464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Grundhoff A, Ganem D. Mechanisms governing expression of the v-FLIP gene of Kaposi’s sarcoma-associated herpesvirus. J Virol. 2001;75(4):1857–1863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Cai X, Cullen BR. Transcriptional origin of Kaposi’s sarcoma-associated herpesvirus microRNAs. J Virol. 2006;80(5):2234–2242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Samols MA, Hu J, Skalsky RL, Renne R. Cloning and identification of a microRNA cluster within the latency-associated region of Kaposi’s sarcoma-associated herpesvirus. J Virol. 2005;79(14):9301–9305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Cai X, Lu S, Zhang Z, Gonzalez CM, Damania B, Cullen BR. Kaposi’s sarcoma-associated herpesvirus expresses an array of viral microRNAs in latently infected cells. Proc Natl Acad Sci U S A. 2005;102(15):5570–5575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Forte E, Raja AN, Shamulailatpam P, et al. MicroRNA-mediated transformation by the Kaposi’s sarcoma-associated herpesvirus Kaposin locus. J Virol. 2015;89(4):2333–2341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Staskus KA, Zhong W, Gebhard K, et al. Kaposi’s sarcoma-associated herpesvirus gene expression in endothelial (spindle) tumor cells. J Virol. 1997;71(1):715–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.O’Hara AJ, Chugh P, Wang L, et al. Pre-micro RNA signatures delineate stages of endothelial cell transformation in Kaposi sarcoma. PLoS Pathog. 2009;5(4):e1000389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Rose TM, Bruce AG, Barcy S, et al. Quantitative RNAseq analysis of Ugandan KS tumors reveals KSHV gene expression dominated by transcription from the LTd downstream latency promoter. PLoS Pathog. 2018;14(12):e1007441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Dittmer DP. Transcription profile of Kaposi’s sarcoma-associated herpesvirus in primary Kaposi’s sarcoma lesions as determined by real-time PCR arrays. Cancer Res. 2003;63(9):2010–2015. [PubMed] [Google Scholar]
  • 81.Arias C, Weisburd B, Stern-Ginossar N, et al. KSHV 2.0: a comprehensive annotation of the Kaposi’s sarcoma-associated herpesvirus genome using next-generation sequencing reveals novel genomic and functional features. PLoS Pathog. 2014;10(1):e1003847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Majerciak V, Ni T, Yang W, Meng B, Zhu J, Zheng ZM. A viral genome landscape of RNA polyadenylation from KSHV latent to lytic infection. PLoS Pathog. 2013;9(11):e1003749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.McClure LV, Kincaid RP, Burke JM, Grundhoff A, Sullivan CS. Comprehensive mapping and analysis of Kaposi’s sarcoma-associated herpesvirus 3’ UTRs identify differential posttranscriptional control of gene expression in lytic versus latent infection. J Virol. 2013;87(23):12838–12849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ballestas ME, Chatis PA, Kaye KM. Efficient persistence of extrachromosomal KSHV DNA mediated by latency-associated nuclear antigen. Science. 1999;284(5414):641–644. [DOI] [PubMed] [Google Scholar]
  • 85.Barbera AJ, Chodaparambil JV, Kelley-Clarke B, et al. The nucleosomal surface as a docking station for Kaposi’s sarcoma herpesvirus LANA. Science. 2006;311(5762):856–861. [DOI] [PubMed] [Google Scholar]
  • 86.Godfrey A, Anderson J, Papanastasiou A, Takeuchi Y, Boshoff C. Inhibiting primary effusion lymphoma by lentiviral vectors encoding short hairpin RNA. Blood. 2005;105(6):2510–2518. [DOI] [PubMed] [Google Scholar]
  • 87.Hilton IB, Simon JM, Lieb JD, Davis IJ, Damania B, Dittmer DP. The open chromatin landscape of Kaposi’s sarcoma-associated herpesvirus. J Virol. 2013;87(21):11831–11842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Toth Z, Maglinte DT, Lee SH, et al. Epigenetic analysis of KSHV latent and lytic genomes. PLoS pathogens. 2010;6(7):e1001013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Gunther T, Grundhoff A. The epigenetic landscape of latent Kaposi sarcoma-associated herpesvirus genomes. PLoS Pathog. 2010;6(6):e1000935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Gunther T, Frohlich J, Herrde C, et al. A comparative epigenome analysis of gammaherpesviruses suggests cis-acting sequence features as critical mediators of rapid polycomb recruitment. PLoS Pathog. 2019;15(10):e1007838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Parravicini C, Chandran B, Corbellino M, et al. Differential viral protein expression in Kaposi’s sarcoma-associated herpesvirus-infected diseases: Kaposi’s sarcoma, primary effusion lymphoma, and multicentric Castleman’s disease. Am J Pathol. 2000;156(3):743–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Aoki Y, Yarchoan R, Wyvill K, Okamoto S, Little RF, Tosato G. Detection of viral interleukin-6 in Kaposi sarcoma-associated herpesvirus-linked disorders. Blood. 2001;97(7):2173–2176. [DOI] [PubMed] [Google Scholar]
  • 93.Abere B, Mamo TM, Hartmann S, et al. The Kaposi’s sarcoma-associated herpesvirus (KSHV) non-structural membrane protein K15 is required for viral lytic replication and may represent a therapeutic target. PLoS Pathog. 2017;13(9):e1006639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Poole LJ, Zong JC, Ciufo DM, et al. Comparison of genetic variability at multiple loci across the genomes of the major subtypes of Kaposi’s sarcoma-associated herpesvirus reveals evidence for recombination and for two distinct types of open reading frame K15 alleles at the right-hand end. J Virol. 1999;73(8):6646–6660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Brinkmann MM, Glenn M, Rainbow L, Kieser A, Henke-Gendo C, Schulz TF. Activation of mitogen-activated protein kinase and NF-kappaB pathways by a Kaposi’s sarcoma-associated herpesvirus K15 membrane protein. J Virol. 2003;77(17):9346–9358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Chugh PE, Sin SH, Ozgur S, et al. Systemically circulating viral and tumor-derived microRNAs in KSHV-associated malignancies. PLoS Pathog. 2013;9(7):e1003484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.McNamara RP, Chugh PE, Bailey A, et al. Extracellular vesicles from Kaposi Sarcoma-associated herpesvirus lymphoma induce long-term endothelial cell reprogramming. PLoS Pathog. 2019;15(2):e1007536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Hansen A, Henderson S, Lagos D, et al. KSHV-encoded miRNAs target MAF to induce endothelial cell reprogramming. Genes Dev. 2010;24(2):195–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Naipauer J, Rosario S, Gupta S, et al. PDGFRA defines the mesenchymal stem cell Kaposi’s sarcoma progenitors by enabling KSHV oncogenesis in an angiogenic environment. PLoS Pathog. 2019;15(12):e1008221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Naipauer J, Salyakina D, Journo G, et al. High-throughput sequencing analysis of a “hit and run” cell and animal model of KSHV tumorigenesis. PLoS Pathog. 2020;16(6):e1008589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Ma Q, Cavallin LE, Yan B, et al. Antitumorigenesis of antioxidants in a transgenic Rac1 model of Kaposi’s sarcoma. Proc Natl Acad Sci U S A. 2009;106(21):8683–8688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Mutlu AD, Cavallin LE, Vincent L, et al. In vivo-restricted and reversible malignancy induced by human herpesvirus-8 KSHV: a cell and animal model of virally induced Kaposi’s sarcoma. Cancer Cell. 2007;11(3):245–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Sturzl M, Blasig C, Schreier A, et al. Expression of HHV-8 latency-associated T0.7 RNA in spindle cells and endothelial cells of AIDS-associated, classical and African Kaposi’s sarcoma. Int J Cancer. 1997;72(1):68–71. [DOI] [PubMed] [Google Scholar]
  • 104.Dupin N, Fisher C, Kellam P, et al. Distribution of human herpesvirus-8 latently infected cells in Kaposi’s sarcoma, multicentric Castleman’s disease, and primary effusion lymphoma. Proc Natl Acad Sci U S A. 1999;96(8):4546–4551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Krown SE, Metroka C, Wernz JC. Kaposi’s sarcoma in the acquired immune deficiency syndrome: a proposal for uniform evaluation, response, and staging criteria. AIDS Clinical Trials Group Oncology Committee. J Clin Oncol. 1989;7(9):1201–1207. [DOI] [PubMed] [Google Scholar]
  • 106.Hosseinipour MC, Kang M, Krown SE, et al. As-Needed Vs Immediate Etoposide Chemotherapy in Combination With Antiretroviral Therapy for Mild-to-Moderate AIDS-Associated Kaposi Sarcoma in Resource-Limited Settings: A5264/AMC-067 Randomized Clinical Trial. Clin Infect Dis. 2018;67(2):251–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Burger H, Ismail Z, Taljaard JJ. Establishing a multidisciplinary AIDS-associated Kaposi’s sarcoma clinic: Patient characteristics, management and outcomes. S Afr Med J. 2018;108(12):1059–1065. [DOI] [PubMed] [Google Scholar]
  • 108.Stallone G, Schena A, Infante B, et al. Sirolimus for Kaposi’s sarcoma in renal-transplant recipients. N Engl J Med. 2005;352(13):1317–1323. [DOI] [PubMed] [Google Scholar]
  • 109.Krown SE, Roy D, Lee JY, et al. Rapamycin with antiretroviral therapy in AIDS-associated Kaposi sarcoma: an AIDS Malignancy Consortium study. J Acquir Immune Defic Syndr. 2012;59(5):447–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Koon HB, Bubley GJ, Pantanowitz L, et al. Imatinib-induced regression of AIDS-related Kaposi’s sarcoma. J Clin Oncol. 2005;23(5):982–989. [DOI] [PubMed] [Google Scholar]
  • 111.Uldrick TS, Goncalves PH, Wyvill KM, et al. A Phase Ib Study of Sorafenib (BAY 43–9006) in Patients with Kaposi Sarcoma. The oncologist. 2017;22(5):505–e549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Reid EG, Suazo A, Lensing SY, et al. Pilot Trial AMC-063: Safety and Efficacy of Bortezomib in AIDS-associated Kaposi Sarcoma. Clin Cancer Res. 2020;26(3):558–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Hong YK, Foreman K, Shin JW, et al. Lymphatic reprogramming of blood vascular endothelium by Kaposi sarcoma-associated herpesvirus. Nat Genet. 2004;36(7):683–685. [DOI] [PubMed] [Google Scholar]
  • 114.Wang HW, Trotter MW, Lagos D, et al. Kaposi sarcoma herpesvirus-induced cellular reprogramming contributes to the lymphatic endothelial gene expression in Kaposi sarcoma. Nat Genet. 2004;36(7):687–693. [DOI] [PubMed] [Google Scholar]
  • 115.Wigle JT, Harvey N, Detmar M, et al. An essential role for Prox1 in the induction of the lymphatic endothelial cell phenotype. EMBO J. 2002;21(7):1505–1513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Weninger W, Partanen TA, Breiteneder-Geleff S, et al. Expression of vascular endothelial growth factor receptor-3 and podoplanin suggests a lymphatic endothelial cell origin of Kaposi’s sarcoma tumor cells. Lab Invest. 1999;79(2):243–251. [PubMed] [Google Scholar]
  • 117.Tuohinto K, DiMaio TA, Kiss EA, et al. KSHV infection of endothelial precursor cells with lymphatic characteristics as a novel model for translational Kaposi’s sarcoma studies. PLoS Pathog. 2023;19(1):e1010753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Liu R, Li X, Tulpule A, et al. KSHV-induced notch components render endothelial and mural cell characteristics and cell survival. Blood. 2010;115(4):887–895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Cheng F, Pekkonen P, Laurinavicius S, et al. KSHV-initiated notch activation leads to membrane-type-1 matrix metalloproteinase-dependent lymphatic endothelial-to-mesenchymal transition. Cell Host Microbe. 2011;10(6):577–590. [DOI] [PubMed] [Google Scholar]
  • 120.Gasperini P, Espigol-Frigole G, McCormick PJ, et al. Kaposi sarcoma herpesvirus promotes endothelial-to-mesenchymal transition through Notch-dependent signaling. Cancer Res. 2012;72(5):1157–1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Li Y, Zhong C, Liu D, et al. Evidence for Kaposi Sarcoma Originating from Mesenchymal Stem Cell through KSHV-induced Mesenchymal-to-Endothelial Transition. Cancer Research. 2018;78(1):230–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Cavallin LE, Ma Q, Naipauer J, et al. KSHV-induced ligand mediated activation of PDGF receptor-alpha drives Kaposi’s sarcomagenesis. PLoS Pathog. 2018;14(7):e1007175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Parsons CH, Szomju B, Kedes DH. Susceptibility of human fetal mesenchymal stem cells to Kaposi sarcoma-associated herpesvirus. Blood. 2004;104(9):2736–2738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Lee MS, Yuan H, Jeon H, et al. Human Mesenchymal Stem Cells of Diverse Origins Support Persistent Infection with Kaposi’s Sarcoma-Associated Herpesvirus and Manifest Distinct Angiogenic, Invasive, and Transforming Phenotypes. MBio. 2016;7(1):e02109–02115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Carroll PA, Brazeau E, Lagunoff M. Kaposi’s sarcoma-associated herpesvirus infection of blood endothelial cells induces lymphatic differentiation. Virology. 2004;328(1):7–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Gramolelli S, Elbasani E, Tuohinto K, et al. Oncogenic Herpesvirus Engages Endothelial Transcription Factors SOX18 and PROX1 to Increase Viral Genome Copies and Virus Production. Cancer Res. 2020;80(15):3116–3129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Ding Y, Chen W, Lu Z, Wang Y, Yuan Y. Kaposi’s sarcoma-associated herpesvirus promotes mesenchymal-to-endothelial transition by resolving the bivalent chromatin of PROX1 gene. PLoS Pathog. 2021;17(9):e1009847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Morris VA, Punjabi AS, Wells RC, Wittkopp CJ, Vart R, Lagunoff M. The KSHV viral IL-6 homolog is sufficient to induce blood to lymphatic endothelial cell differentiation. Virology. 2012;428(2):112–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Sodhi A, Chaisuparat R, Hu J, et al. The TSC2/mTOR pathway drives endothelial cell transformation induced by the Kaposi’s sarcoma-associated herpesvirus G protein-coupled receptor. Cancer Cell. 2006;10(2):133–143. [DOI] [PubMed] [Google Scholar]
  • 130.Montaner S, Sodhi A, Molinolo A, et al. Endothelial infection with KSHV genes in vivo reveals that vGPCR initiates Kaposi’s sarcomagenesis and can promote the tumorigenic potential of viral latent genes. Cancer Cell. 2003;3(1):23–36. [DOI] [PubMed] [Google Scholar]
  • 131.Yogev O, Lagos D, Enver T, Boshoff C. Kaposi’s sarcoma herpesvirus microRNAs induce metabolic transformation of infected cells. PLoS Pathog. 2014;10(9):e1004400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Boss IW, Nadeau PE, Abbott JR, Yang Y, Mergia A, Renne R. A Kaposi’s sarcoma-associated herpesvirus-encoded ortholog of microRNA miR-155 induces human splenic B-cell expansion in NOD/LtSz-scid IL2Rgammanull mice. J Virol. 2011;85(19):9877–9886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Jones T, Ye F, Bedolla R, et al. Direct and efficient cellular transformation of primary rat mesenchymal precursor cells by KSHV. J Clin Invest. 2012;122(3):1076–1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Friborg J, Jr., Kong W, Hottiger MO, Nabel GJ. p53 inhibition by the LANA protein of KSHV protects against cell death. Nature. 1999;402(6764):889–894. [DOI] [PubMed] [Google Scholar]
  • 135.Douglas JL, Whitford JG, Moses AV. Characterization of c-Kit expression and activation in KSHV-infected endothelial cells. Virology. 2009;390(2):174–185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.McHugh D, Caduff N, Barros MHM, et al. Persistent KSHV Infection Increases EBV-Associated Tumor Formation In Vivo via Enhanced EBV Lytic Gene Expression. Cell Host Microbe. 2017;22(1):61–73 e67. [DOI] [PubMed] [Google Scholar]
  • 137.Ito Y, Hart JR, Ueno L, Vogt PK. Oncogenic activity of the regulatory subunit p85beta of phosphatidylinositol 3-kinase (PI3K). Proc Natl Acad Sci U S A. 2014;111(47):16826–16829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.ar-Rushdi A, Nishikura K, Erikson J, Watt R, Rovera G, Croce CM. Differential expression of the translocated and the untranslocated c-myc oncogene in Burkitt lymphoma. Science. 1983;222(4622):390–393. [DOI] [PubMed] [Google Scholar]
  • 139.Rabbitts TH, Hamlyn PH, Baer R. Altered nucleotide sequences of a translocated c-myc gene in Burkitt lymphoma. Nature. 1983;306(5945):760–765. [DOI] [PubMed] [Google Scholar]
  • 140.Mesri EA, Feitelson MA, Munger K. Human viral oncogenesis: a cancer hallmarks analysis. Cell Host Microbe. 2014;15(3):266–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Ang KK, Harris J, Wheeler R, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 2010;363(1):24–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Halfon P, Berger P, Khiri H, et al. Algorithm based on CMV kinetics DNA viral load for preemptive therapy initiation after hematopoietic cell transplantation. J Med Virol. 2011;83(3):490–495. [DOI] [PubMed] [Google Scholar]
  • 143.Gulley ML, Glaser SL, Craig FE, et al. Guidelines for interpreting EBER in situ hybridization and LMP1 immunohistochemical tests for detecting Epstein-Barr virus in Hodgkin lymphoma. Am J Clin Pathol. 2002;117(2):259–267. [DOI] [PubMed] [Google Scholar]
  • 144.Kanakry JA, Hegde AM, Durand CM, et al. The clinical significance of EBV DNA in the plasma and peripheral blood mononuclear cells of patients with or without EBV diseases. Blood. 2016;127(16):2007–2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Decker LL, Shankar P, Khan G, et al. The Kaposi sarcoma-associated herpesvirus (KSHV) is present as an intact latent genome in KS tissue but replicates in the peripheral blood mononuclear cells of KS patients. J Exp Med. 1996;184(1):283–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Suresh R, Mosser DM. Pattern recognition receptors in innate immunity, host defense, and immunopathology. Adv Physiol Educ. 2013;37(4):284–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Frazao A, Rethacker L, Messaoudene M, et al. NKG2D/NKG2-Ligand Pathway Offers New Opportunities in Cancer Treatment. Front Immunol. 2019;10:661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Moore PS, Chang Y. Kaposi’s sarcoma-associated herpesvirus immunoevasion and tumorigenesis: two sides of the same coin? Annu Rev Microbiol. 2003;57:609–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Broussard G, Damania B. KSHV: Immune Modulation and Immunotherapy. Front Immunol. 2019;10:3084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Uppal T, Sarkar R, Dhelaria R, Verma SC. Role of Pattern Recognition Receptors in KSHV Infection. Cancers (Basel). 2018;10(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.West J, Damania B. Upregulation of the TLR3 pathway by Kaposi’s sarcoma-associated herpesvirus during primary infection. J Virol. 2008;82(11):5440–5449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Jacobs SR, Damania B. The viral interferon regulatory factors of KSHV: immunosuppressors or oncogenes? Front Immunol. 2011;2:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Meyer W, Liumsiricharoen M, Suprasert A, Fleischer LG, Hewicker-Trautwein M. Immunohistochemical demonstration of keratins in the epidermal layers of the Malayan pangolin (Manis javanica), with remarks on the evolution of the integumental scale armour. Eur J Histochem. 2013;57(3):e27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Lingel A, Ehlers E, Wang Q, et al. Kaposi’s Sarcoma-Associated Herpesvirus Reduces Cellular Myeloid Differentiation Primary-Response Gene 88 (MyD88) Expression via Modulation of Its RNA. J Virol. 2016;90(1):180–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Gregory SM, Davis BK, West JA, et al. Discovery of a viral NLR homolog that inhibits the inflammasome. Science. 2011;331(6015):330–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Yang X, Zhou J, Liu C, et al. KSHV-encoded ORF45 activates human NLRP1 inflammasome. Nat Immunol. 2022;23(6):916–926. [DOI] [PubMed] [Google Scholar]
  • 157.West JA, Wicks M, Gregory SM, et al. An important role for mitochondrial antiviral signaling protein in the Kaposi’s sarcoma-associated herpesvirus life cycle. J Virol. 2014;88(10):5778–5787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Zhao Y, Ye X, Dunker W, Song Y, Karijolich J. RIG-I like receptor sensing of host RNAs facilitates the cell-intrinsic immune response to KSHV infection. Nat Commun. 2018;9(1):4841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Zhang H, Ni G, Damania B. ADAR1 Facilitates KSHV Lytic Reactivation by Modulating the RLR-Dependent Signaling Pathway. Cell Rep. 2020;31(4):107564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Zhang Y, Dittmer DP, Mieczkowski PA, et al. RIG-I Detects Kaposi’s Sarcoma-Associated Herpesvirus Transcripts in a RNA Polymerase III-Independent Manner. MBio. 2018;9(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Inn KS, Lee SH, Rathbun JY, et al. Inhibition of RIG-I-mediated signaling by Kaposi’s sarcoma-associated herpesvirus-encoded deubiquitinase ORF64. J Virol. 2011;85(20):10899–10904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Wu JJ, Li W, Shao Y, et al. Inhibition of cGAS DNA Sensing by a Herpesvirus Virion Protein. Cell Host Microbe. 2015;18(3):333–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Zhang G, Chan B, Samarina N, et al. Cytoplasmic isoforms of Kaposi sarcoma herpesvirus LANA recruit and antagonize the innate immune DNA sensor cGAS. Proc Natl Acad Sci U S A. 2016;113(8):E1034–1043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Ma Z, Jacobs SR, West JA, et al. Modulation of the cGAS-STING DNA sensing pathway by gammaherpesviruses. Proc Natl Acad Sci U S A. 2015;112(31):E4306–4315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Manes TD, Hoer S, Muller WA, Lehner PJ, Pober JS. Kaposi’s sarcoma-associated herpesvirus K3 and K5 proteins block distinct steps in transendothelial migration of effector memory CD4+ T cells by targeting different endothelial proteins. J Immunol. 2010;184(9):5186–5192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Nachmani D, Stern-Ginossar N, Sarid R, Mandelboim O. Diverse Herpesvirus MicroRNAs Target the Stress-Induced Immune Ligand MICB to Escape Recognition by Natural Killer Cells. Cell Host & Microbe. 2009;5(4):376–385. [DOI] [PubMed] [Google Scholar]
  • 167.Sun Z, Jha HC, Pei YG, Robertson ES. Major Histocompatibility Complex Class II HLA-DRalpha Is Downregulated by Kaposi’s Sarcoma-Associated Herpesvirus-Encoded Lytic Transactivator RTA and MARCH8. J Virol. 2016;90(18):8047–8058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Santarelli R, Granato M, Faggioni A, Cirone M. Interference with the Autophagic Process as a Viral Strategy to Escape from the Immune Control: Lesson from Gamma Herpesviruses. J Immunol Res. 2015;2015:546063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Schmidt K, Wies E, Neipel F. Kaposi’s sarcoma-associated herpesvirus viral interferon regulatory factor 3 inhibits gamma interferon and major histocompatibility complex class II expression. J Virol. 2011;85(9):4530–4537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Cai Q, Banerjee S, Cervini A, et al. IRF-4-Mediated CIITA Transcription Is Blocked by KSHV Encoded LANA to Inhibit MHC II Presentation. PLOS Pathogens. 2013;9(10):e1003751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Pontejo SM, Murphy PM. Chemokines encoded by herpesviruses. J Leukoc Biol. 2017;102(5):1199–1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Yamin R, Kaynan NS, Glasner A, et al. The viral KSHV chemokine vMIP-II inhibits the migration of Naive and activated human NK cells by antagonizing two distinct chemokine receptors. PLoS Pathog. 2013;9(8):e1003568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Cousins E, Gao Y, Sandford G, Nicholas J. Human herpesvirus 8 viral interleukin-6 signaling through gp130 promotes virus replication in primary effusion lymphoma and endothelial cells. J Virol. 2014;88(20):12167–12172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Li X, Bhaduri-McIntosh S. A Central Role for STAT3 in Gammaherpesvirus-Life Cycle and -Diseases. Front Microbiol. 2016;7:1052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Rivera-Soto R, Damania B. Modulation of Angiogenic Processes by the Human Gammaherpesviruses, Epstein-Barr Virus and Kaposi’s Sarcoma-Associated Herpesvirus. Front Microbiol. 2019;10:1544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Kang S, Tanaka T, Narazaki M, Kishimoto T. Targeting Interleukin-6 Signaling in Clinic. Immunity. 2019;50(4):1007–1023. [DOI] [PubMed] [Google Scholar]
  • 177.Chen D, Gao Y, Nicholas J. Human herpesvirus 8 interleukin-6 contributes to primary effusion lymphoma cell viability via suppression of proapoptotic cathepsin D, a cointeraction partner of vitamin K epoxide reductase complex subunit 1 variant 2. J Virol. 2014;88(2):1025–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Rosario SA, Santiago GE, Mesri EA, Verdun RE. Kaposi’s Sarcoma-Associated Herpesvirus-Encoded Viral IL-6 (vIL-6) Enhances Immunoglobulin Class-Switch Recombination. Front Microbiol. 2018;9:3119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Yu L, Tu M, Cortes J, et al. Clinical and pathological characteristics of HIV- and HHV-8-negative Castleman disease. Blood. 2017;129(12):1658–1668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Ramaswami R, Lurain K, Peer CJ, et al. Tocilizumab in patients with symptomatic Kaposi sarcoma herpesvirus-associated multicentric Castleman disease. Blood. 2020;135(25):2316–2319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Host KM, Jacobs SR, West JA, et al. Kaposi’s Sarcoma-Associated Herpesvirus Increases PD-L1 and Proinflammatory Cytokine Expression in Human Monocytes. MBio. 2017;8(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Beldi-Ferchiou A, Lambert M, Dogniaux S, et al. PD-1 mediates functional exhaustion of activated NK cells in patients with Kaposi sarcoma. Oncotarget. 2016;7(45):72961–72977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Galanina N, Goodman AM, Cohen PR, Frampton GM, Kurzrock R. Successful Treatment of HIV-Associated Kaposi Sarcoma with Immune Checkpoint Blockade. Cancer Immunol Res. 2018;6(10):1129–1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Uldrick TS, Goncalves PH, Hay MMA, et al. Phase I study of pembrolizumab in people with HIV and cancer. Journal of Clinical Oncology. 2019;37(15_suppl):2500–2500. [Google Scholar]
  • 185.Uldrick TS, Goncalves PH, Abdul-Hay M, et al. Assessment of the Safety of Pembrolizumab in Patients With HIV and Advanced Cancer-A Phase 1 Study. JAMA Oncol. 2019;5(9):1332–1339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Delyon J, Biard L, Renaud M, et al. PD-1 blockade with pembrolizumab in classic or endemic Kaposi’s sarcoma: a multicentre, single-arm, phase 2 study. Lancet Oncol. 2022;23(4):491–500. [DOI] [PubMed] [Google Scholar]
  • 187.Rasmussen TA, Rajdev L, Rhodes A, et al. Impact of Anti-PD-1 and Anti-CTLA-4 on the Human Immunodeficiency Virus (HIV) Reservoir in People Living With HIV With Cancer on Antiretroviral Therapy: The AIDS Malignancy Consortium 095 Study. Clin Infect Dis. 2021;73(7):e1973–e1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Uldrick TS, Adams SV, Fromentin R, et al. Pembrolizumab induces HIV latency reversal in people living with HIV and cancer on antiretroviral therapy. Sci Transl Med. 2022;14(629):eabl3836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Lingel H, Brunner-Weinzierl MC. CTLA-4 (CD152): A versatile receptor for immune-based therapy. Semin Immunol. 2019;42:101298. [DOI] [PubMed] [Google Scholar]
  • 190.Tabata MM, Novoa RA, Bui NQ, Zaba LC. Successful treatment of HIV-negative Kaposi sarcoma with ipilimumab and nivolumab and concurrent management of baseline psoriasis and bullous pemphigoid. JAAD Case Rep. 2020;6(5):447–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Zer A, Icht O, Yosef L, et al. Phase II single-arm study of nivolumab and ipilimumab (Nivo/Ipi) in previously treated classical Kaposi sarcoma (cKS). Ann Oncol. 2022;33(7):720–727. [DOI] [PubMed] [Google Scholar]
  • 192.Ramaswami R, Polizzotto MN, Lurain K, et al. Safety, activity, and long-term outcomes of pomalidomide in the treatment of Kaposi sarcoma among individuals with or without HIV infection. Clin Cancer Res. 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Polizzotto MN, Uldrick TS, Wyvill KM, et al. Pomalidomide for Symptomatic Kaposi’s Sarcoma in People With and Without HIV Infection: A Phase I/II Study. J Clin Oncol. 2016;34(34):4125–4131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Ramaswami R, Polizzotto MN, Lurain K, et al. Safety, Activity, and Long-term Outcomes of Pomalidomide in the Treatment of Kaposi Sarcoma among Individuals with or without HIV Infection. Clin Cancer Res. 2022;28(5):840–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Patil A, Manzano M, Gottwein E. Genome-wide CRISPR screens reveal genetic mediators of cereblon modulator toxicity in primary effusion lymphoma. Blood Adv. 2019;3(14):2105–2117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Shrestha P, Davis DA, Jaeger HK, Stream A, Aisabor AI, Yarchoan R. Pomalidomide restores immune recognition of primary effusion lymphoma through upregulation of ICAM-1 and B7–2. PLoS Pathog. 2021;17(1):e1009091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Davis DA, Mishra S, Anagho HA, et al. Restoration of immune surface molecules in Kaposi sarcoma-associated herpes virus infected cells by lenalidomide and pomalidomide. Oncotarget. 2017;8(31):50342–50358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Fujiwara Y, Sun Y, Torphy RJ, et al. Pomalidomide Inhibits PD-L1 Induction to Promote Antitumor Immunity. Cancer Res. 2018;78(23):6655–6665. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

As this is a review, the data are available in PubMed and as listed in references.

RESOURCES