Skip to main content
Elsevier Sponsored Documents logoLink to Elsevier Sponsored Documents
. 2018 Oct;32:60–70. doi: 10.1016/j.coviro.2018.08.014

Contribution of the KSHV and EBV lytic cycles to tumourigenesis

Oliver Manners 1,2, James C Murphy 1,2, Alex Coleman 1,2, David J Hughes 3, Adrian Whitehouse 1,2,4
PMCID: PMC6259586  PMID: 30268927

Abstract

Kaposi’s Sarcoma-associated herpesvirus (KSHV) and Epstein Barr virus (EBV) are the causative agents of several malignancies. Like all herpesviruses, KSHV and EBV undergo distinct latent and lytic replication programmes. The transition between these states allows the establishment of a lifelong persistent infection, dissemination to sites of disease and the spread to new hosts. Latency-associated viral proteins have been well characterised in transformation and tumourigenesis pathways; however, a number of studies have shown that abrogation of KSHV and EBV lytic gene expression impairs the oncogenesis of several cancers. Furthermore, several lytically expressed proteins have been functionally tethered to the angioproliferative and anti-apoptotic phenotypes of virus-infected cells. As a result, the investigation and therapeutic targeting of KSHV and EBV lytic cycles may be essential for the treatment of their associated malignancies.


Current Opinion in Virology 2018, 32:60–70

This review comes from a themed issue on Viruses and cancer

Edited by Cary Moody and Erle Robertson

For a complete overview see the Issue and the Editorial

Available online 28th September 2018

https://doi.org/10.1016/j.coviro.2018.08.014

1879-6257/© 2018 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

Introduction

Kaposi’s sarcoma associated herpesvirus (KSHV) and Epstein Barr virus (EBV) are double stranded gammaherpesviruses which contribute to the oncogenesis of several human tumours. KSHV is the etiological agent of the endothelial cell tumour Kaposi’s Sarcoma, in addition to two lymphoproliferative disorders; primary effusion lymphoma (PEL) and multicentric Castleman’s disease (MCD) [1, 2, 3]. Whereas, EBV has been linked with multiple malignancies including Burkitt’s lymphoma (BL), Hodgkin’s lymphoma (HL), nasopharyngeal carcinoma (NPC) and gastric carcinoma (GC) [4, 5, 6].

Like all herpesviruses, KSHV and EBV have a biphasic life cycle comprising latent and lytic replication programmes. During latency, both viruses exist in a dormant state where only a subset of the viral genes are expressed facilitating the episomal persistence of the viral genome [7,8]. However, under certain physiological conditions, both viruses undergo lytic reactivation leading to expression of the full complement of lytic genes followed by the assembly and egress of infectious virions. Importantly however, both KSHV and EBV can also undergo abortive lytic reactivation, resulting in the expression of early lytic genes without subsequent virion assembly and cell lysis.

Although much of the efforts to understand the molecular basis of these disorders has focused on viral latency, KSHV and EBV lytic cycles are now widely accepted as major contributors to oncogenesis which could be important targets in the development of anti-cancer therapeutics [9,10]. Thus, in this review, we discuss how lytic replication augments the pathogenesis of KSHV and EBV-associated malignancies (Figure 1) and the treatments available which may target the lytic replication cycle.

Figure 1.

Figure 1

Schematic representation how KSHV and EBV lytically expressed proteins augment the pathogenesis of KSHV and EBV-associated malignancies.

KSHV lytic factors and tumourigenesis

Expression of the KSHV major lytic transactivator, RTA, is sufficient and necessary to activate the KSHV lytic cycle leading to a triphasic transcriptional cascade of immediate early, delayed early and late gene expression [11,12]. To initiate the transition from latency to lytic replication, a range of stimuli have been implicated including hypoxia, co-infection with HIV-1, oxidative stress and inflammatory cytokines; all of which promote the expression of RTA [13, 14, 15, 16]. Importantly, the treatment of KS patients with drugs that prevent lytic replication can, in certain cases, lead to regression of KS lesions; attesting to the importance of lytic gene expression in tumourigenesis [17].

Although in vivo, the spontaneous reactivation of KSHV takes place in only 1–3% of infected cells, the resultant assembly and egress of KSHV infectious particles sustains the population of latently infected cells that would otherwise be lost due to a combination of defects in episome synthesis during cell division and the death of infected cells [18]. Therefore, the balance between KSHV latent and lytic replication programmes is stringently controlled to ensure viral persistence and consequently tumour development. Nevertheless, in addition to the role of the lytic cycle in supporting a lifelong, persistent KSHV infection, a number of lytic factors themselves have oncogenic properties [19] (Table 1). Thus, the following section will outline these oncogenic lytic genes and discuss how they contribute to malignancy alongside their role in viral replication.

Table 1.

KSHV lytic oncogenes and mechanisms of tumorigenesis

KSHV lytic gene Cellular homologue Lytic function Mechanisms of oncogenesis Oncogenic function Reference
vGPRCR IL-8 receptor Activates cellular signalling pathways to maintain Rta expression Secretes paracrine signalling molecules such as VEGF Cell survival, angiogenesis [22, 23, 24, 25,28]
Activates Rho and Rac1 GTPases
Stimulates PI3K, PDK, AKT/PKB, p38 and MAPK signalling pathways
K1 BCR Activates cellular signalling pathways to maintain Rta expression Contributes to VEGF autocrine and paracrine signalling Cell survival, angiogenesis [31, 32, 33, 34, 35,36]
Perturbs normal PI3K and MAPK signalling
Activates AKT and mTOR
Interacts with HIV-1 Tat to activate NF-κB
K15 Activates cellular signalling pathways Modulates PLCγ-Calcineurin-NFAT pathways Angiogenesis [37,38]
Chemokine and cytokine production
vIL-6 IL-6 Immune evasion and activation of cellular signalling pathways Activates JAK/STAT, MAPK and PI3K/AKT pathways Proliferation and angiogenesis [40, 41, 42, 43]
Contributes to VEGF autocrine and paracrine signalling
vIRFs IRFs Immunoevasins Inhibits Interferon α/β/γ responses and inflammatory signalling Cell survival [39,44, 45, 46, 47]
Interaction with p53 prevents ATM/p53 DNA damage response pathway
vBCL-2 BCL-2 Delays cell death Inhibits apoptosis and autophagy Cell survival [48, 49, 50,51,52]
vCCLs CCLs Immunoevasins Modulation of CCL activity Angiogenesis [53, 54, 55]
Chemoattractants for TH2 cells to reduce TH1 cell activity
RTA Lytic transactivator Induction of DNA damage Genome instability [60]
ORF57 Processing, export and translation of viral RNAs Sequesters the transcription and export complex (hTREX) to cause R-loop formation and DSBs Genome instability [57,58,59]

vGPCR

Viral G-protein-coupled-receptor (vGPCR) is an early lytic protein encoded by the viral gene ORF74. It shares limited homology to the human interleukin-8 receptors CXCR1 and CXCR2; however, ORF74 is constitutively active and minimally responsive to various ligands [20,21]. vGPCR activates members of the G protein family which stimulate several major cell signalling pathways such as phosphatidylinositol 3-kinase (PI3K), phosphoinoside-dependent kinase (PDK) and AKT/protein kinase B (AKT/PKB) along with several small GTPase proteins such as RhoA and Rac1 [22, 23, 24, 25]. The induction of a myriad of signalling pathways which activate Sp1/3 transcription factors is crucial to the maintenance of RTA expression and therefore commits the cell to productive lytic replication [26,27]. Previous studies have demonstrated that vGPCR is an oncogene capable of inducing angioproliferative lesions in transgenic mice that bear the pathological indicators of KS [28]. The angioproliferative nature of vGPCR appears to stem from its indirect production and secretion of a number of paracrine signalling molecules such as vascular endothelial growth factor (VEGF) [22, 23, 24, 25]. As a result, it has been suggested that vGPCR initiates the immortalisation of endothelial cells and KS tumourigenesis through the establishment of a VEGF paracrine loop [28]. In this proposed ‘hit and run’ model of immortalisation, populations of lytically replicating endothelial cells express vGPCR leading to VEGF expression and secretion. This acts in an autocrine mechanism on lytically replicating cells but also in a paracrine fashion on adjacent latently infected cells to promote their survival and immortalisation.

K1 and K15

ORF-K1 encodes a type 1 transmembrane glycoprotein which shares structural similarities with the B cell receptor (BCR) [29]. The lytic protein appears to contribute to KSHV lytic replication in a similar manner to vGPCR through the maintenance of RTA expression; however, the specific mechanism behind this is not well understood [30]. Like vGPCR, K1 expression also contributes to the VEGF autocrine and paracrine signalling loop through its constitutively active ITAM domain, promoting the production and secretion of VEGF via matrix metalloprotease 9 (MMP-9) [31, 32, 33]. Increased VEGF signalling modulates downstream pathways such as PI3K and mitogen activated kinase which in turn activate Akt kinase and mammalian target of rapamycin (mTOR) to promote cell survival [32,34,35]. Furthermore, K1 has also been shown to interact with the HIV-1 protein Tat. Mouse studies have shown that coexpression of K1 and HIV-1 Tat leads to a synergistic increase in angiogenesis through the Tat-mediated upregulation of host microRNA miR-891a-5b which targets NF-κβ [36].

Like ORF-K1, KSHV ORF-K15 encodes a transmembrane receptor which antagonises BCR signalling; however, K15 lack an ITAM domain and activates the production of cytokines and chemokines via cellular signalling pathways including NF-κB, MEK/ERK and Jun N-terminal protein kinase (JNK) pathways [37]. K15 bypasses the induction of VEGF signalling by activating the downstream PLCγ-Calcineurin-NFAT pathways to facilitate angiogenic tube formation in cell culture [38]. It is speculated that K15 is involved in the early development of KS tumour where limited lytic gene expression is detected [38].

KSHV immunoevasins

Successful KSHV lytic replication is dependent on the expression of viral interferon regulatory factors (IRFs) and viral interleukins (IL) which prevent immune detection [39]. Most of these viral proteins are expressed during lytic replication to inhibit the interferon (IFN) antiviral response. KSHV vIL-6, like its human homologue can bind the gp130 receptor and activate the JAK/STAT, MAPK and PI3K/Akt cellular signalling pathways leading to expression of hIL-6 and VEGF [40, 41, 42]. Furthermore, the inoculation of NIH3T3 cells ectopically expressing vIL-6 into immunocompromised mice leads to tumour formation and vIL-6 expression in endothelial cells leads to angioproliferation and tubule formation [43].

The KSHV genome also possesses three lytically encoded vIRF ORFs: ORF-K9, ORF-K11/K11.1 and ORF-K10 encoding vIRF1, vIRF2 and vIRF4 respectively [39,44]. vIRFs act to disrupt the antiviral IFN response by inhibiting transcription of IFN-α/-β/-γ and inflammatory signals. The mechanism of vIRF action varies between isoforms; however, in many cases vIRF bind to cellular IRFs and inhibit their ability to activate transcription [39]. Through the dysregulation of the IFN antiviral response, apoptosis and cell cycle arrest are prevented increasing the oncogenic potential of KSHV-infected cells [44,45]. vIRF1 co-precipitates with p53, reducing p53 target gene expression [46]. Studies have demonstrated this vIRF1-p53 is crucial for the inhibition of the ATM/p53 DNA damage response pathway allowing viral DNA replication to proceed [39,46,47]. vIRF1 appears to block activation of ATM and decreases p53 stability through reduced phosphorylation of Ser15 of p53 [47].

vBcl-2

vBcl-2, encoded by KSHV ORF16 shares sequence and functional homology to the Bcl-2 family of cellular proteins [48]. Normally, Bcl-2 proteins act as regulators of apoptosis and are characterised by four conserved stretches of amino acids called Bcl-2 homology (BH) domains [49]. Cellular Bcl-2 also negatively regulates autophagy by interacting with autophagy promoting factor Beclin-2 [50]. Studies in the murine counterpart of KSHV, murine herpesvirus 68 (MHV68), have shown that vBcl-2 is involved in both the inhibition of autophagy and apoptosis to promote B cell survival [51]. The importance of autophagy in programmed cell death makes it an important target for KSHV during lytic infection to prevent induction of apoptosis and thus the impairment of viral replication. In KSHV the function of vBcl-2 has not yet been fully elucidated; however, lack of vBcl-2 impairs KSHV reactivation [48,52].

vCCLs

KSHV also encodes three homologues of cellular chemokines known as viral CC-chemokine ligands (vCCLs) [53]. Previous studies have shown that vCCL1–vCCL3 are all able to bind to their cellular homologues both agonistically and antagonistically to stimulate angiogenesis [53,54]. Furthermore, all three viral proteins have been suggested to act as chemoattractants to modulate the levels of different T-cell subpopulations in KS lesions. The resultant TH2 cell-predominant tumour microenvironment in which TH1 cell responses are downregulated allowing immune evasion and thus tumour progression [55].

KSHV and genome instability

KSHV lytic replication is associated with the formation of double strand-breaks (DSBs) and chromosomal aberrations, which are a common feature of KS lesions [56]. The KSHV early protein ORF57 controls the processing, export and translation of viral RNAs [57,58]. However, studies have shown that sequestration of the human transcription and export complex (hTREX) by ORF57 leads to DSBs as a result of R-loop formation [59]. Furthermore, studies have suggested that Rta is also able to induce DNA damage [60]. Importantly, the activation of members of the DNA damage response machinery in this way has been suggested to facilitate viral DNA synthesis during productive lytic replication; as is the case with other Herpesviruses [60]. Thus, the role of KSHV lytic factors in genome instability likely contributes to oncogenesis.

EBV lytic factors and tumourigenesis

EBV establishes a lifelong infection in B lymphocytes achieved through a highly regulated viral gene expression program. In latently infected B cells, the expression of either Zta or Rta is sufficient to reactivate the EBV lytic cycle [61, 62, 63, 64]. Lytic replication can be studied by treating latently infected B cells with inducers of the lytic cycle such as phorbal esters or by crosslinking B cell receptors with anti-immunoglobulins [65,66]. There have been several important insights regarding the contribution of the EBV lytic cycle for virus-induced tumorigenesis in vivo. Mouse models have demonstrated that lytic replication incompetent-EBV particles are impaired in their ability to cause lymphomagenesis compared to wild type virus, despite similar infection levels [67]. Furthermore, acyclovir treatment, which blocks lytic viral genome replication but not lytic gene expression, is unable to prevent EBV associated lymphomagenesis; reinforcing the role of lytic cycle-induced paracrine signals in disease progression [68]. Finally, in vivo studies have shown that in KSHV-infected PEL cells, tumour formation is enhanced upon coinfection with EBV [69••]. Together, these studies strongly support the view that lytic gene expression is important for tumour progression, and that paracrine signals play an essential role.

Similar to KSHV and the role of lytic reactivation in KS, reactivation of EBV may aid transmission of the virus within the tumor microenvironment to establish latency and drive cellular proliferation. However, the likely predominant role of the EBV lytic cycle is to provide the necessary paracrine, anti-apoptotic and immunomodulatory signals required for tumorigenesis (Table 2).

Table 2.

EBV lytic oncogenes and mechanisms of tumorigenesis

EBV lytic gene Cellular homologue Lytic function Mechanisms of oncogenesis Oncogenic function Reference
ZTA Lytic transactivator Promotes secretion of proangiogenic factors IL-6, IL-8, IL-10, IL-13 and VEGF Angiogenesis [67,68,70,71]
BHRF1 and BALF1 BCL-2 Delay cell death Inhibition of apoptosis Cell survival [73,74]
BILF1 Immunoevasin Unknown but detected in EBV tumours [82,83]
BNFR1 Nucleocapsid trafficking Interacts with centromeres to cause centrosome overduplication Genome instability [85••]
BALF3 Terminase DNA synthesis and incorporation into virons DNA damage Genome instability [88]
BGLF4 Viral DNA replication and nuclear import of viral proteins DNA damage Genome instability [86]
BGLF5 Host cell shut-off DNA damage Genome instability [79, 80, 81,87]
BARF1 C-fms receptor Immunomodulator Modulates expression genes involved in apoptotic signalling Cell survival [76,77]
BCRF1 IL-10 Immunoevasin Downregulates interferon γ Cell survival [75]

ZTA

Various studies have shown that the expression of some lytic antigens alone is sufficient to induce the expression of immunomodulatory and paracrine factors associated with oncogenesis. The lytic transactivator Zta facilitates the secretion of IL-6, IL-8, IL-10 and IL-13 in addition to proangiogenic proteins such as vascular endothelial growth factor (VEGF) [67,68,70]. Soluble Zta has also been detected in the sera of post-transplant lymphoproliferative disease (PTLD) patients providing further evidence of a transformative role for this lytic protein [70]. Finally, Zta alone can downregulate the expression of CIITA, an essential transcription factor important for HLA-II expression permitting immune evasion and tumour progression [71].

EBV and anti-apoptotic signalling

In addition to the paracrine effect, some lytic proteins elicit strong anti-apoptotic signals [72]. Like KSHV vBcl-2, the EBV lytic cycle-associated proteins BHRF1 and BALF1 are viral homologs of cellular Bcl-2 which perform anti-apoptotic functions critical for cellular transformation in vitro [73,74]. Similarly, BCRF1, which is analogous to cellular IL-10, increases the viability and transformation of EBV-infected B cells through downregulation of interferon-γ [75]. Finally, BARF1 is one of the most highly expressed genes in NPC cell lines and antibodies are frequently detected in NPC-patient sera [73]. The encoded protein, BARF1, a homolog of colony-stimulating factor 1 receptor, is a secreted anti-apoptotic factor which influences the survival of neighbouring cells [76,77]. Taken together, these studies implicate anti-apoptotic signalling by EBV lytic proteins in the oncogenesis of EBV-associated malignancies. However, although the expression of BHRF1, BALF1 and BARF1 is dramatically increased during lytic reactivation, and they have previously been designated as lytic genes, their expression has been detected in LCLs (lymphoblastoid cell lines) where cells are predominantly latently infected (>95%) [78]. Furthermore, BHRF1 is expressed from the latent promoter Wp in a subset of BL known as Wp-restricted BL [78]. Therefore, it is unclear whether these proteins contribute to tumorigenesis during the latent or lytic life cycles.

BGLF5 and BILF1 immunoevasins

Several EBV lytic proteins, which primarily function as immunoevasins, also contribute to tumorigenesis. BGLF5, the EBV host shut-off protein which inhibits translation of host mRNAs, functions in the downregulation of toll-like receptor 9 (TLR9) and human leukocyte antigen class I (HLA-I) and –II leading to impaired T cell recognition [79,80]. Importantly however, BGLF5 expression has been detected in NPC biopsies and BGLF5 antibodies have been detected in NPC patient sera, suggesting the protein also undertakes a transformative role [81]. Similarly, the lytic EBV immunoevasin BILF1 enhances the internalisation of surface molecules of HLA-I leading to their rapid degradation by the lysosome; again impairing T cell recognition [82]. However, BILF1 expression has also been detected in NPC cells, once again suggesting an oncogenic function [83].

EBV and genomic instability

Like KSHV, several EBV lytic proteins have also been implicated as contributors to genomic instability [84]. A recent study suggested that the EBV major tegument protein, BNFR1, involved in translocation of the viral nucleocapsid to the nucleus, induces centrosome amplification and thus contributes to the accumulation of chromosomal aberrations in infected cells [85••]. Furthermore, BNFR1 was able to induce genomic instability within latently infected LCLs without necessarily establishing infection, demonstrating that EBV lytic replication can promote transformation in adjacent cells [85••]. Three further EBV proteins, BALF3, BGLF4 and host cell-shutoff protein BGLF5 have all been suggested to further contribute to chromosomal instability and tumourigenesis through induction of DNA damage in NPC cells [86, 87, 88]. Thus, genomic instability appears a major mechanism through which EBV lytic proteins contribute to oncogenesis.

Treatments for KSHV and EBV-associated cancers

Considerable advances in the understanding of the KSHV and EBV life cycle and related pathologies have been made since their discovery, however, presently there are still no vaccines or effective direct therapeutic options available for the prevention or treatment of their associated cancers. Almost all clinically available therapies that target the lytic life cycle of KSHV and EBV do not directly inhibit the virus and have shown varying results in the clinic (Table 3). Because of the critical role of lytic replication in disease progression and virus dissemination these highlight the need for potent and selective therapeutics against lytic viral targets to treat KSHV and EBV-associated cancers.

Table 3.

Current treatments and inhibitors targeting the lytic life cycles of KSHV and EBV-associated cancers. Stage of development abbreviations: R = Randomised, C = Control and SP = Single Patient. Clinical disease abbreviations: HSV = Herpes simplex virus, VZV = Varicella zoster virus, CMV = Cytomegalovirus, HBV = Hepatitis B virus, ML = Myeloid leukaemia, ALL = Acute lymphoblastic leukaemia, LAM = Lymphangioleiomyomatosis, MDS = Myelodysplastic syndromes, CMML = Chronic myelomonocytic leukaemia and CTCL = Cutaneous T-cell lymphoma

Therpay target Inhibitor class Inhibitor subclass Drug Stage of development
Already clinically approved for:
KSHV EBV
Viral DNA polymerase Nucleoside analogues Purine analogues Acyclovir Cohort study In vitro HSV and VZV
Valganciclovir R, C trial SP study CMV
Pyrimidine analogs Zidovudine R trial In vivo HIV
Methylenecyclopropane nucleosides Cyclopropavir In vitro In vitro None
Acyclic nucleoside phosphonates HPMP derivatives Cidofovir Pilot study In vivo CMV
PME derivatives Adefovir In vitro In vitro HBV
Pyrophosphate analog Foscarnet sodium Cohort study In vitro CMV
Non-nucleoside inhibitor Pyrimidoquinoline analog NSC 373989 In vitro None None
Viral mRNAs Peptide-conjugated phosphorodiamidate morpholin oligomers (PPMO) PPMO against RTA In vivo n/a None
PPMO against vIRF-1 In vitro n/a None
Viral capsid protease Small molecule helical mimetic Dimerisation inhibitor DD2 In vitro In vitro None
Lytic repression Small-molecule inhibitors Stilbenoid Resveratrol In vitro In vitro Dietary supplement
Lipid Delta-9 tetrahydrocannabinol In vitro In vitro Multiple sclerosis
Ephrin receptor tyrosine kinase A2 Small-molecule inhibitors ATP-competitive tyrosine kinase inhibitor Dasatinib In vitro Phase I trial Chronic ML and ALL
4-(2,5-dimethyl-pyrrol-1-yl)-2-hydroxy-benzoic acid In vitro None None
Dihydroorotate dehydrogenase/p53 Small-molecule inhibitor Teriflunomide None In vitro Multiple sclerosis
HSP70 Small-molecule inhibitor ATP-derivative inhibitor VER-155008 In vitro None None
hTREX Small-molecule inhibitor ATPase inhibitor CCT018159 In vitro None None
mTOR Polyketide Rapamycin Phase I trial In vivo Organ transplant rejection and LAM
Lytic induction Cytidine analogue DNA demethylation agent 5-azacytidine In vitro In vitro MDS, Acute ML and CMML
Small-molecule inhibitor N-protected dipeptide Bortezomib In vivo In vitro Multiple myeloma
Neddylation inhibitor Pevonedistat In vitro In vitro None
Lipid Valproic acid In vitro Pilot study Anticonvulsant
Histone deacetylase inhibitor Suberoylanilide hydroxamic acid In vivo In vivo CTCL
C7 None In vitro None
Protective T-cell immunity Vaccine Capsid protein gp350 n/a Phase II trial None
Replication-competent viruses Latency deficient replication-competent viruses In vivo None None
Cytokine therapy GM-CSF and IL-2 None In vivo None

There is no standard of care for the treatment of KSHV-associated tumours and current options range from targeting cancers through surgical excision, chemotherapy and radiotherapy [89]. These treatment options are also recommended for EBV-associated cancers however, due to the array of cancers associated with EBV, treatment guidelines vary greatly for each different associated cancer [90].

To date, the most effective treatment of AIDS-related KS and AIDS-related EBV-associated cancers is highly active antiretroviral therapy (HAART), which works mostly through restoration of the patient’s immune system [91]. Likewise, iatrogenic KS, MCD, PEL and iatrogenic EBV-associated sarcomas and lymphomas are treated through the removal of immunosuppressants, to restore the patient’s immune system, limiting tumour progression, however this in turn can lead to graft rejection [92].

Immunotherapies have also been demonstrated to be effective at treating KSHV and EBV-associated cancers. Rituximab (anti-CD20) is clinically approved for the treatment of many EBV-associated lymphoproliferative diseases and also MCD [93••,94]. In addition, Tocilizumab (anti-human IL-6 receptor) and Siltuximab (anti-IL6 chimeric monoclonal antibody) are clinically available for the treatment of MCD [95,96].

Novel therapies involving lytic KSHV and EBV

The majority of cells present in KSHV and EBV-associated cancers are latently infected therefore, there is substantial research exploring the potential of lytic induction therapy to treat these cancers. This treatment involves the efficient induction of all latently infected tumour cells into the lytic cycle while concomitantly exposing the cells to inhibitors of lytic replication and inducing apoptosis to clear all virally infected cells. In addition, lytic induction therapy can help induce a cytotoxic T-lymphocyte (CTL) response to lytic antigens to further clear cancerous virally infected cells. Lytic induction therapy poses a powerful mechanism to enhance the efficacy of EBV and KSHV lytic inhibitors, which are discussed below.

Because of the critical role of the KSHV and EBV lytic life cycle in tumorigenesis there is considerable interest in developing vaccines that target lytic antigens [97]. Various vaccines have been created targeting a range of lytic KSHV and EBV antigens. Some of these vaccines have proved successful in vivo and in clinical trials, such as the EBV envelope protein gp350, which reduced primary infection however, the vaccine failed to decrease the overall EBV infection rate.

Finally, cytokine therapy can induced protective T-cell immunity against viruses. Cytokine therapy in a humanised mouse model with EBV-associated lymphoproliferative disease induced a marked expansion of Zta-specific T-cells, which can prolong survival [98,99]. In addition to supporting the notion that Zta plays a critical role in lymphoproliferative disease, it provides a strong rationale for the inclusion of Zta-specific antigens in vaccine development [97].

KSHV and EBV lytic inhibitors

The lytic life cycles of KSHV and EBV pose numerous attractive and viable targets for the development of anti-viral drugs. However, the only KSHV and EBV inhibitors clinically available (and all anti-herpesvirus drugs in general) target the viral DNA-polymerase [100]. The most common of these drugs are nucleoside analogues, but acyclic nucleoside phosphonates (ANPs) and pyrophosphate analogues are also frequently used for herpesvirus therapy [89]. Clinically available nucleoside analogues are administered as pro-drugs and only activated by the viral thymidine kinase present during lytic replication [101]. Novel viral DNA-polymerase inhibitors have also been developed such as, second and third generation, nucleoside analogues and ANPs, and non-nucleoside inhibitors [102,103].

Many other inhibitors of herpesvirus replication have also been explored which target numerous aspects of the lytic life cycle, however none have made it as yet into the clinic. Targets with inhibitors demonstrated to have efficacy against EBV and KSHV include, the KSHV latent-lytic transactivator RTA, KSHV IRFs and the viral capsid protease [104,105].

Cellular targets required for lytic reactivation of EBV and KSHV and that contribute to lytic EBV and KSHV-associated tumorigenesis have also been approached as targets for inhibitors of their associated cancers [106]. Cellular inhibitors bear an increased risk of cytotoxic side effects, however, they possess great advantages due to fewer occurrences of drug-resistance and the potential for a broader activity against a range of viruses. Targets include, the KSHV/EBV cellular entry receptor (ephrin receptor tyrosine kinase A2), the proteasome, Hsp70, the human transcription/export complex (hTREX), the mammalian target of rapamycin (mTOR) and the dihydroorotate dehydrogenase enzyme [106,107,108,109,110,111]. While most inhibitors of cellular targets have only been demonstrated in vitro some have also shown efficacy in the clinic.

All clinically available inhibitors of EBV and KSHV have low efficacy and only target the viral DNA polymerase. Therefore, more efforts should be invested to examine the potential of drugs that target other viral proteins, since this proof-of-principle has been shown beneficial for other herpesviruses, such as HSV and HCMV.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as

  • • of special interest

  • •• of outstanding interest

Acknowledgements

Research in the Whitehouse laboratory is supported in parts by funding from Biotechnology and Biological Sciences Research Council (BB/M006557/1, BB/N014405/1), Medical Research Council (MR/R010145/1, 95505168), Worldwide Cancer Research (16-1025), Wellcome Trust (203826/Z/16/Z) and Rosetrees Trust (M662).

References

  • 1.Chang Y., Cesarman E., Pessin M.S., Lee F., Culpepper J., Knowles D.M., Moore P.S. Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi’s sarcoma. Science. 1994;266:1865–1869. doi: 10.1126/science.7997879. [DOI] [PubMed] [Google Scholar]
  • 2.Cesarman E., Chang Y., Moore P.S., Said J.W., Knowles D.M. Kaposi’s sarcoma–associated herpesvirus-like DNA sequences in AIDS-related body-cavity–based lymphomas. N Engl J Med. 1995;332:1186–1191. doi: 10.1056/NEJM199505043321802. [DOI] [PubMed] [Google Scholar]
  • 3.Soulier J., Grollet L., Oksenhendler E., Cacoub P., Cazals-Hatem D., Babinet P., D’Agay M.F., Clauvel J.P., Raphael M., Degos L. Kaposi’s sarcoma-associated herpesvirus-like DNA sequences in multicentric Castleman’s disease. Blood. 1995;86:1276–1280. [PubMed] [Google Scholar]
  • 4.Coates P.J., Slavin G., D’Ardenne A.J. Persistence of Epstein-Barr virus in Reed-Sternberg cells throughout the course of Hodgkin’s disease. J Pathol. 1991;164:291–297. doi: 10.1002/path.1711640404. [DOI] [PubMed] [Google Scholar]
  • 5.de Schryver A., Friberg S.J., Klein G., Henle W., Henle G., De-The G., Clifford P., Ho H.C. Epstein-Barr virus-associated antibody patterns in carcinoma of the post-nasal space. Clin Exp Immunol. 1969;5:443–459. [PMC free article] [PubMed] [Google Scholar]
  • 6.Hzur Hausen, Schulte-Holthausen H., Klein G., Henle W., Henle G., Clifford P., Santesson L. Epstein–Barr virus in Burkitts lymphoma and nasopharyngeal carcinoma: EBV DNA in biopsies of Burkitt tumours and anaplastic carcinomas of the nasopharynx. Nature. 1970;228:1056. doi: 10.1038/2281056a0. [DOI] [PubMed] [Google Scholar]
  • 7.Kenney S.C., Mertz J.E. Regulation of the latent-lytic switch in Epstein–Barr virus. Semin Cancer Biol. 2014;0:60–68. doi: 10.1016/j.semcancer.2014.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cai Q., Verma S.C., Lu J., Robertson E.S. Molecular biology of Kaposi's sarcoma herpesvirus and related oncogenesis. Adv Virus Res. 2010;78:87–142. doi: 10.1016/B978-0-12-385032-4.00003-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hardie D.R. Human gamma-herpesviruses: a review of 2 divergent paths to oncogenesis. Transfus Apher Sci. 2010;42:177–183. doi: 10.1016/j.transci.2010.01.015. [DOI] [PubMed] [Google Scholar]
  • 10.Jha H., Banerjee S., Robertson E. The role of gammaherpesviruses in cancer pathogenesis. Pathogens. 2016;5:18. doi: 10.3390/pathogens5010018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lukac D.M., Kirshner J.R., Ganem D. Transcriptional activation by the product of open reading frame 50 of Kaposi’s sarcoma-associated herpesvirus is required for lytic viral reactivation in B cells. J Virol. 1999;73:9348–9361. doi: 10.1128/jvi.73.11.9348-9361.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wilson S.J., Tsao E.H., Webb B.L.J., Ye H., Dalton-Griffin L., Tsantoulas C., Gale C.V., Du M.-Q., Whitehouse A., Kellam P. X box binding protein XBP-1s transactivates the Kaposi’s sarcoma-associated herpesvirus (KSHV) ORF50 promoter, linking plasma cell differentiation to KSHV reactivation from latency. J Virol. 2007;81:13578–13586. doi: 10.1128/JVI.01663-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Davis D.A., Rinderknecht A.S., Zoeteweij J.P., Aoki Y., Read-connole E.L., Tosato G., Blauvelt A., Yarchoan R. Hypoxia induces lytic replication of Kaposi sarcoma-associated herpesvirus. Vascular. 2011;97:3244–3250. doi: 10.1182/blood.v97.10.3244. [DOI] [PubMed] [Google Scholar]
  • 14.Cohen A., Brodie C., Sarid R. An essential role of ERK signalling in TPA-induced reactivation of Kaposi’s sarcoma-associated herpesvirus. J Gen Virol. 2006;87:795–802. doi: 10.1099/vir.0.81619-0. [DOI] [PubMed] [Google Scholar]
  • 15.Merat R., Amara A., Lebbe C., De The H., Morel P., Saib A. HIV-1 infection of primary effusion lymphoma cell line triggers Kaposi’s sarcoma-associated herpesvirus (KSHV) reactivation. Int J Cancer. 2002;97:791–795. doi: 10.1002/ijc.10086. [DOI] [PubMed] [Google Scholar]
  • 16.Mercader M., Taddeo B., Panella J.R., Chandran B., Nickoloff B.J., Foreman K.E. Induction of HHV-8 lytic cycle replication by inflammatory cytokines produced by HIV-1-infected T cells. Am J Pathol. 2000;156:1961–1971. doi: 10.1016/S0002-9440(10)65069-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Martin J.N., Osmond D.H. Kaposi’s sarcoma-associated herpesvirus and sexual transmission of cancer risk. Curr Opin Oncol. 1999;11 doi: 10.1097/00001622-199911000-00013. [DOI] [PubMed] [Google Scholar]
  • 18.Chiu Y.F., Sugden A.U., Fox K., Hayes M., Sugden B. Kaposi’s sarcoma-associated herpesvirus stably clusters its genomes across generations to maintain itself extrachromosomally. J Cell Biol. 2017;216:2745–2758. doi: 10.1083/jcb.201702013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Wen K.W., Damania B. Kaposi Sarcoma-associated Herpesvirus (KSHV): molecular biology and oncogenesis. Cancer Lett. 2010;289:140–150. doi: 10.1016/j.canlet.2009.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Guo H.-G., Browning P., Nicholas J., Hayward G.S., Tschachler E., Jang Y.-W., Sadowski M., Raffeld M., Colombini S., Gallo R.C. Characterization of a chemokine-related gene in human herpesvirus 8 and its expression in Kaposi’s sarcoma. Virology. 1996;228:371. doi: 10.1006/viro.1996.8386. [DOI] [PubMed] [Google Scholar]
  • 21.Rosenkilde M.M., Kledal T.N., Bräuner-Osborne H., Schwartz T.W. Agonists and inverse agonists for the herpesvirus 8-encoded constitutively active seven-transmembrane oncogene product, ORF-74. J Biol Chem. 1999;274:956–961. doi: 10.1074/jbc.274.2.956. [DOI] [PubMed] [Google Scholar]
  • 22.Sodhi A., Montaner S., Patel V., Zohar M., Bais C., Mesri E.A., Gutkind J.S. The Kaposi’s sarcoma-associated herpes virus G protein-coupled receptor up-regulates vascular endothelial growth factor expression and secretion through mitogen-activated protein kinase and p38 pathways acting on hypoxia-inducible factor 1alpha. Cancer Res. 2000;60:4873–4880. [PubMed] [Google Scholar]
  • 23.Bais C., Van Geelen A., Eroles P., Mutlu A., Chiozzini C., Dias S., Silverstein R.L., Rafii S., Mesri E.A. Kaposi’s sarcoma associated herpesvirus G protein-coupled receptor immortalizes human endothelial cells by activation of the VEGF receptor-2/ KDR. Cancer Cell. 2003;3:131–143. doi: 10.1016/s1535-6108(03)00024-2. [DOI] [PubMed] [Google Scholar]
  • 24.Montaner S., Sodhi A., Pece S., Mesri E.A., Gutkind J.S. The Kaposi’s sarcoma-associated herpesvirus G protein-coupled receptor promotes endothelial cell survival through the activation of Akt/protein kinase B. Cancer Res. 2001;61:2641–2648. [PubMed] [Google Scholar]
  • 25.Martín M.J., Tanos T., García A.B., Martin D., Gutkind J.S., Coso O.A., Marinissen M.J. The Galpha12/13 family of heterotrimeric G proteins and the small GTPase RhoA link the Kaposi sarcoma-associated herpes virus G protein-coupled receptor to heme oxygenase-1 expression and tumorigenesis. J Biol Chem. 2007;282:34510–34524. doi: 10.1074/jbc.M703043200. [DOI] [PubMed] [Google Scholar]
  • 26.Chiou C.-J., Poole L.J., Kim P.S., Ciufo D.M., Cannon J.S., ap Rhys C.M., Alcendor D.J., Zong J.-C., Ambinder R.F., Hayward G.S. Patterns of gene expression and a transactivation function exhibited by the vGCR (ORF74) chemokine receptor protein of Kaposi’s sarcoma-associated herpesvirus. J Virol. 2002;76:3421–3439. doi: 10.1128/JVI.76.7.3421-3439.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bottero V., Sharma-Walia N., Kerur N., Paul A.G., Cannon M., Chandran B. Kaposi sarcoma- associated herpes virus (KSHV) G protein-coupled receptor (vGPCR) activates the ORF50 lytic switch promoter: a potential positive feedback loop for sustained ORF50 gene expression. Virology. 2010;392:34–51. doi: 10.1016/j.virol.2009.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bais C., Santomasso B., Coso O., Arvanitakis L., Raaka E.G., Gutkind J.S., Asch A.S., Cesarman E., Gerhengorn M.C., Mesri E.A. G-protein-coupled receptor of Kaposi’s sarcoma-associated herpesvirus is a viral oncogene and angiogenesis activator. Nature. 1998;391:86–89. doi: 10.1038/34193. [DOI] [PubMed] [Google Scholar]
  • 29.Steinbrück L., Gustems M., Medele S., Schulz T.F., Lutter D., Hammerschmidt W. K1 and K15 of Kaposi’s sarcoma-associated herpesvirus are partial functional homologues of latent membrane protein 2A of Epstein-Barr virus. J Virol. 2015;89:7248–7261. doi: 10.1128/JVI.00839-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Zhang Z., Chen W., Sanders M.K., Brulois K.F., Dittmer D.P., Damania B. The K1 protein of Kaposi’s sarcoma-associated herpesvirus (KSHV) augments viral lytic replication. J Virol. 2016;90:JVI.03102–03115. doi: 10.1128/JVI.03102-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Belotti D., Paganoni P., Manenti L., Garofalo A., Marchini S., Taraboletti G., Giavazzi R. Matrix metalloproteinases (MMP9 and MMP2) induce the release of vascular endothelial growth factor (VEGF) by ovarian carcinoma cells: implications for ascites formation. Cancer Res. 2003;63:5224–5229. [PubMed] [Google Scholar]
  • 32.Lagunoff M., Majeti R., Weiss A., Ganem D. Deregulated signal transduction by the K1 gene product of Kaposi’s sarcoma-associated herpesvirus. Proc Natl Acad Sci U S A. 1999;96:5704–5709. doi: 10.1073/pnas.96.10.5704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lee H., Guo J., Li M., Choi J.K., DeMaria M., Rosenzweig M., Jung J.U. Identification of an immunoreceptor tyrosine-based activation motif of K1 transforming protein of Kaposi’s sarcoma-associated herpesvirus. Mol Cell Biol. 1998;18:5219–5228. doi: 10.1128/mcb.18.9.5219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Brinkmann M., Glenn M., Rainbow L., Kieser A., Henke-Gendo C., Schulz T.F. Activation of mitogen-activated protein kinase and NF-kappaB pathways by a Kaposi’s sarcoma-associated herpesvirus K15 membrane protein. J Virol. 2003;77:9346–9358. doi: 10.1128/JVI.77.17.9346-9358.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Wang L., Dittmer D.P., Tomlinson C.C., Fakhari F.D., Damania B. Immortalization of primary endothelial cells by the K1 protein of Kaposi’s sarcoma-associated herpesvirus. Cancer Res. 2006;66:3658–3666. doi: 10.1158/0008-5472.CAN-05-3680. [DOI] [PubMed] [Google Scholar]
  • 36•.Yao S., Hu M., Hao T., Li W., Xue X., Xue M., Zhu X., Zhou F., Qin D., Yan Q. MiRNA-891a-5p mediates HIV-1 Tat and KSHV Orf-K1 synergistic induction of angiogenesis by activating NF-κB signaling. Nucleic Acids Res. 2015;43:9362–9378. doi: 10.1093/nar/gkv988. [DOI] [PMC free article] [PubMed] [Google Scholar]; This report demonstrates that HIV-1 protein Tat promotes KSHV K1-induced angiogenesis through the upregulation of a microRNAs which in turn activates NF-κB signalling.
  • 37.Brinkmann M.M., Pietrek M., Dittrich-Breiholz O., Kracht M., Schulz T.F. Modulation of host gene expression by the K15 protein of Kaposi’s sarcoma-associated herpesvirus. J Virol. 2007;81:42–58. doi: 10.1128/JVI.00648-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bala K., Bosco R., Gramolelli S., Haas D.A., Kati S., Pietrek M., Hävemeier A., Yakushko Y., Singh V.V., Dittrich-Breiholz O. Kaposi’s sarcoma herpesvirus K15 protein contributes to virus-induced angiogenesis by recruiting PLCγ1 and activating NFAT1-dependent RCAN1 expression. PLoS Pathog. 2012;8 doi: 10.1371/journal.ppat.1002927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Jacobs S.R., Damania B. The viral interferon regulatory factors of KSHV: immunosuppressors or oncogenes. Front Immunol. 2011;2:1–11. doi: 10.3389/fimmu.2011.00019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Osborne J., Moore P.S., Chang Y. KSHV-encoded viral IL-6 activates multiple human IL-6 signaling pathways. Hum Immunol. 1999;60:921–927. doi: 10.1016/s0198-8859(99)00083-x. [DOI] [PubMed] [Google Scholar]
  • 41.An J., Lichtenstein A.K., Brent G., Rettig M.B., An J., Lichtenstein A.K., Brent G., Rettig M.B. The Kaposi sarcoma − associated herpesvirus (KSHV) induces cellular interleukin 6 expression: role of the KSHV latency-associated nuclear antigen and the AP1 response element. Blood. 2002;99:649–654. doi: 10.1182/blood.v99.2.649. [DOI] [PubMed] [Google Scholar]
  • 42.Molden J., Chang Y., You Y., Moore P.S., Goldsmith M.A. A Kaposi’s sarcoma-associated herpesvirus-encoded cytokine homolog (vIL- 6) activates signaling through the shared gp130 receptor subunit. J Biol Chem. 1997;272:19625–19631. doi: 10.1074/jbc.272.31.19625. [DOI] [PubMed] [Google Scholar]
  • 43.Aoki Y., Jaffe E.S., Chang Y., Jones K., Teruya-Feldstein J., Moore P.S., Tosato G. Angiogenesis and hematopoiesis induced by Kaposi’s sarcoma-associated herpesvirus-encoded interleukin-6. Blood. 1999;93:4034–4043. [PubMed] [Google Scholar]
  • 44.Lin R., Genin P., Mamane Y., Sgarbanti M., Battistini A., Harrington W.J., Barber G.N., Hiscott J. HHV-8 encoded vIRF-1 represses the interferon antiviral response by blocking IRF-3 recruitment of the CBP/p300 coactivators. Oncogene. 2001;20:800–811. doi: 10.1038/sj.onc.1204163. [DOI] [PubMed] [Google Scholar]
  • 45.Burýsek L., Yeow W.S., Lubyová B., Kellum M., Schafer S.L., Huang Y.Q., Pitha P.M. Functional analysis of human herpesvirus 8-encoded viral interferon regulatory factor 1 and its association with cellular interferon regulatory factors and p300. J Virol. 1999;73:7334–7342. doi: 10.1128/jvi.73.9.7334-7342.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Seo T., Park J., Lee D., Hwang S.G., Choe J. Viral interferon regulatory factor 1 of Kaposi’s sarcoma-associated herpesvirus binds to p53 and represses p53-dependent transcription and apoptosis. J Virol. 2001;75:6193–6198. doi: 10.1128/JVI.75.13.6193-6198.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Nakamura H., Li M., Zarycki J., Jung J.U. Inhibition of p53 tumor suppressor by viral interferon regulatory factor. J Virol. 2001;75:7572–7582. doi: 10.1128/JVI.75.16.7572-7582.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Gelgor A., Kalt I., Bergson S., Brulois K.F., Jung J.U., Sarid R. Viral Bcl-2 encoded by the Kaposi’s sarcoma-associated herpesvirus is vital for virus reactivation. J Virol. 2015;89:5298–5307. doi: 10.1128/JVI.00098-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Skommer J., Wlodkowic D., Deptala A. Larger than life: mitochondria and the Bcl-2 family. Leuk Res. 2007;31:277–286. doi: 10.1016/j.leukres.2006.06.027. [DOI] [PubMed] [Google Scholar]
  • 50.Pattingre S., Tassa A., Qu X., Garuti R., Xiao H.L., Mizushima N., Packer M., Schneider M.D., Levine B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell. 2005;122:927–939. doi: 10.1016/j.cell.2005.07.002. [DOI] [PubMed] [Google Scholar]
  • 51•.Coleman C.B., McGraw J.E., Feldman E.R., Roth A.N., Keyes L.R., Grau K.R., Cochran S.L., Waldschmidt T.J., Liang C., Forrest J.C. A gammaherpesvirus Bcl-2 ortholog blocks B cell receptor-mediated apoptosis and promotes the survival of developing B cells in vivo. PLoS Pathog. 2014;10 doi: 10.1371/journal.ppat.1003916. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study shows that during MHV 68 infection, vBcl-2 expression is important for the survival of developing B cells.
  • 52.Liang Q., Chang B., Lee P., Brulois K.F., Ge J., Shi M., Rodgers M.A., Feng P., Oh B.-H., Liang C. Identification of the essential role of viral Bcl-2 for Kaposi’s sarcoma-associated herpesvirus lytic replication. J Virol. 2015;89:5308–5317. doi: 10.1128/JVI.00102-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Liu C., Okruzhnov Y., Li H., Nicholas J. Human herpesvirus 8 (HHV-8)-encoded cytokines induce expression of and autocrine signaling by vascular endothelial growth factor (VEGF) in HHV-8-infected primary-effusion lymphoma cell lines and mediate VEGF-independent antiapoptotic effects. J Virol. 2001;75:10933–10940. doi: 10.1128/JVI.75.22.10933-10940.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Choi Y.B., Nicholas J. Induction of angiogenic chemokine CCL2 by human herpesvirus 8 chemokine receptor. Virology. 2010;397:369–378. doi: 10.1016/j.virol.2009.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lee H.-R., Lee S., Chaudhary P.M., Gill P., Jung J.U. Immune evasion by Kaposi’s sarcoma-associated herpesvirus. Future Microbiol. 2010;5:1349–1365. doi: 10.2217/fmb.10.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Xiao Y., Chen J., Liao Q., Wu Y., Peng C., Chen X. Lytic infection of Kaposi’s sarcoma-associated herpesvirus induces DNA double-strand breaks and impairs non-homologous end joining. J Gen Virol. 2013;94:1870–1875. doi: 10.1099/vir.0.053033-0. [DOI] [PubMed] [Google Scholar]
  • 57.Jackson B.R., Boyne J.R., Noerenberg M., Taylor A., Hautbergue G.M., Walsh M.J., Wheat R., Blackbourn D.J., Wilson S.A., Whitehouse A. An interaction between KSHV ORF57 and UIF provides mRNA-adaptor redundancy in herpesvirus intronless mRNA export. PLoS Pathog. 2011;7 doi: 10.1371/journal.ppat.1002138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Boyne J.R., Jackson B.R., Taylor A., MacNab S.A., Whitehouse A. Kaposi’s sarcoma-associated herpesvirus ORF57 protein interacts with PYM to enhance translation of viral intronless mRNAs. EMBO J. 2010;29:1851–1864. doi: 10.1038/emboj.2010.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59•.Jackson B.R., Noerenberg M., Whitehouse A. A novel mechanism inducing genome instability in Kaposi’s sarcoma-associated herpesvirus infected cells. PLoS Pathog. 2014;10 doi: 10.1371/journal.ppat.1004098. [DOI] [PMC free article] [PubMed] [Google Scholar]; This report provides evidence that KSHV ORF57 leads to sequestration of the human TREX complex and double strand breaks as a result of R-loop formation.
  • 60.Hollingworth R., Skalka G.L., Stewart G.S., Hislop A.D., Blackbourn D.J., Grand R.J. Activation of DNA damage response pathways during lytic replication of KSHV. Viruses. 2015;7:2908–2927. doi: 10.3390/v7062752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Countryman J., Miller G. Activation of expression of latent Epstein-Barr herpesvirus after gene transfer with a small cloned subfragment of heterogeneous viral DNA. Proc Natl Acad Sci U S A. 1985;82:4085–4089. doi: 10.1073/pnas.82.12.4085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Feederle R. The Epstein-Barr virus lytic program is controlled by the co-operative functions of two transactivators. EMBO J. 2000;19:3080–3089. doi: 10.1093/emboj/19.12.3080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Bhende P.M., Dickerson S.J., Sun X., Feng W.-H., Kenney S.C. X-box-binding protein 1 activates lytic Epstein-Barr virus gene expression in combination with protein kinase D. J Virol. 2007;81:7363–7370. doi: 10.1128/JVI.00154-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Takada K., Shimizu N., Sakuma S., Ono Y. Trans activation of the latent Epstein-Barr virus (EBV) genome after transfection of the EBV DNA fragment. J Virol. 1986;57:1016–1022. doi: 10.1128/jvi.57.3.1016-1022.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Takada K. Cross-linking of cell surface immunoglobulin induces Epstein-Barr virus in Burkitt Lymphoma Lines. Int J Cancer. 1984;33:27–32. doi: 10.1002/ijc.2910330106. [DOI] [PubMed] [Google Scholar]
  • 66.Flemington E., Speck S.H. Identification of phorbol ester response elements in the promoter of Epstein-Barr Virus putative lytic switch gene BZLF1. J Virol. 1990;64:1217–1226. doi: 10.1128/jvi.64.3.1217-1226.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hong G.K., Gulley M.L., Feng W., Delecluse H., Holley-guthrie E., Kenney S.C. Epstein-Barr virus lytic infection contributes to lymphoproliferative disease in a SCID mouse model. J Virol. 2005;79:13993–14003. doi: 10.1128/JVI.79.22.13993-14003.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ma X., Yang L., Xiao L., Tang M., Liu L., Li Z., Deng M., Sun L., Cao Y. Down-regulation of EBV-LMP1 radio-sensitizes nasal pharyngeal carcinoma cells via NF-κB regulated ATM expression. PLoS One. 2011;6 doi: 10.1371/journal.pone.0024647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69••.McHugh D., Caduff N., Barros M.H.M., Rämer P.C., Raykova A., Murer A., Landtwing V., Quast I., Styles C.T., Spohn M. Persistent KSHV infection increases EBV-associated tumor formation in vivo via enhanced EBV lytic gene expression. Cell Host Microbe. 2017;22 doi: 10.1016/j.chom.2017.06.009. 61-73.e7. [DOI] [PubMed] [Google Scholar]; This study demonstrated in vivo persistent KSHV infection enhances EBV lytic gene expression and tumour formation.
  • 70.Katsumura K.R., Maruo S., Takada K. EBV lytic infection enhances transformation of B‐lymphocytes infected with EBV in the presence of T‐lymphocytes. J Med Virol. 2012;84:504–510. doi: 10.1002/jmv.23208. [DOI] [PubMed] [Google Scholar]
  • 71.Balan N., Osborn K., Sinclair A.J. Repression of CIITA by the Epstein-Barr virus transcription factor Zta is independent of its dimerization and DNA binding. J Gen Virol. 2016;97:725–732. doi: 10.1099/jgv.0.000369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Fitzsimmons L., Kelly G.L. EBV and apoptosis: the viral master regulator of cell fate? Viruses. 2017;9:1–34. doi: 10.3390/v9110339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Cabras G., Decaussin G., Zeng Y., Djennaoui D., Melouli H., Broully P., Bouguermouh A.M., Ooka T. Epstein-Barr virus encoded BALF1 gene is transcribed in Burkitt’s lymphoma cell lines and in nasopharyngeal carcinoma’s biopsies. J Clin Virol. 2005;34:26–34. doi: 10.1016/j.jcv.2004.12.016. [DOI] [PubMed] [Google Scholar]
  • 74.Altmann M., Hammerschmidt W. Epstein-Barr virus provides a new paradigm: a requirement for the immediate inhibition of apoptosis. PLoS Biol. 2005;3:1–10. doi: 10.1371/journal.pbio.0030404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Stuart A.D., Stewart J.P., Arrand J.R., Mackett M. The Epstein-Barr virus encoded cytokine viral interleukin-10 enhances transformation of human B lymphocytes. Oncogene. 1995;11:1711–1719. [PubMed] [Google Scholar]
  • 76.Strockbine L.D., Cohen J.I., Farrah T., Lyman S.D., Wagener F., DuBose R.F., Armitage R.J., Spriggs M.K. The Epstein-Barr virus BARF1 gene encodes a novel, soluble colony-stimulating factor-1 receptor. J Virol. 1998;72:4015–4021. doi: 10.1128/jvi.72.5.4015-4021.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hsu W.L., Chung P.J., Tsai M.H., Chang C.L.T., Liang C.L. A role for Epstein-Barr viral BALF1 in facilitating tumor formation and metastasis potential. Virus Res. 2012;163:617–627. doi: 10.1016/j.virusres.2011.12.017. [DOI] [PubMed] [Google Scholar]
  • 78.Kelly G.L., Long H.M., Stylianou J., Thomas W.A., Leese A., Bell A.I., Bornkamm G.W., Mautner J., Rickinson A.B., Rowe M. An Epstein-Barr virus anti-apoptotic protein constitutively expressed in transformed cells and implicated in Burkitt lymphomagenesis: the Wp/BHRF1 link. PLoS Pathog. 2009;5 doi: 10.1371/journal.ppat.1000341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.van Gent M., Griffin B.D., Berkhoff E.G., van Leeuwen D., Boer I.G.J., Buisson M., Hartgers F.C., Burmeister W.P., Wiertz E.J., Ressing M.E. EBV lytic-phase protein BGLF5 contributes to TLR9 downregulation during productive infection. J Immunol. 2011;186:1694–1702. doi: 10.4049/jimmunol.0903120. [DOI] [PubMed] [Google Scholar]
  • 80.Rowe M., Glaunsinger B., van Leeuwen D., Zuo J., Sweetman D., Ganem D., Middeldorp J., Wiertz E.J.H.J., Ressing M.E. Host shutoff during productive Epstein-Barr virus infection is mediated by BGLF5 and may contribute to immune evasion. PNAS. 2007;104:3366–3371. doi: 10.1073/pnas.0611128104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Sbih-Lammali F., Berger F., Busson P., Ooka T. Expression of the DNase encoded by the BGLF5 gene of Epstein-Barr virus in nasopharyngeal carcinoma epithelial cells. Virology. 1996;222:64–74. doi: 10.1006/viro.1996.0398. [DOI] [PubMed] [Google Scholar]
  • 82.Zuo J., Quinn L.L., Tamblyn J., Thomas W.A., Feederle R., Delecluse H.-J., Hislop A.D., Rowe M. The Epstein-Barr virus-encoded BILF1 protein modulates immune recognition of endogenously processed antigen by targeting major histocompatibility complex class I molecules trafficking on both the exocytic and endocytic pathways. J Virol. 2011;85:1604–1614. doi: 10.1128/JVI.01608-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Beisser P., Verzijl D. The Epstein-Barr virus BILF1 gene encodes a G protein-coupled receptor that inhibits phosphorylation of RNA-dependent protein kinase. J Virol. 2005;79:441–449. doi: 10.1128/JVI.79.1.441-449.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Gruhne B., Kamranvar S.A., Masucci M.G., Sompallae R. EBV and genomic instability-a new look at the role of the virus in the pathogenesis of Burkitt’s lymphoma. Semin Cancer Biol. 2009;19:394–400. doi: 10.1016/j.semcancer.2009.07.005. [DOI] [PubMed] [Google Scholar]
  • 85••.Shumilov A., Tsai M.H., Schlosser Y.T., Kratz A.S., Bernhardt K., Fink S., Mizani T., Lin X., Jauch A., Mautner J. Epstein-Barr virus particles induce centrosome amplification and chromosomal instability. Nat Commun. 2017;8:1–15. doi: 10.1038/ncomms14257. [DOI] [PMC free article] [PubMed] [Google Scholar]; This report demonstrates a novel mechanism where EBV particles can induce chromosomal instability in a non-persistent infection. This research reveals a system for development of tumours that do not necessarily carry the EBV genome.
  • 86.Chang Y.H., Lee C.P., Su M.T., Wang J.T., Chen J.Y., Lin S.F., Tsai C.H., Hsieh M.J., Takada K., Chen M.R. Epstein-Barr virus BGLF4 kinase retards cellular S-phase progression and induces chromosomal abnormality. PLoS One. 2012;7 doi: 10.1371/journal.pone.0039217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Wu C.-C., Liu M.-T., Chang Y.-T., Fang C.-Y., Chou S.-P., Liao H.-W., Kuo K.-L., Hsu S.-L., Chen Y.-R., Wang P.-W. Epstein–Barr virus DNase (BGLF5) induces genomic instability in human epithelial cells. Nucleic Acids Res. 2010;38:1932–1949. doi: 10.1093/nar/gkp1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Chiu S.-H., Wu C.-C., Fang C.-Y., Yu S.-L., Hsu H.-Y., Chow Y.-H., Chen J.-Y. Epstein-Barr virus BALF3 mediates genomic instability and progressive malignancy in nasopharyngeal carcinoma. Oncotarget. 2014;5:8583–8601. doi: 10.18632/oncotarget.2323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Coen N., Duraffour S., Snoeck R., Andrei G. KSHV targeted therapy: an update on inhibitors of viral lytic replication. Viruses. 2014;6:4731–4759. doi: 10.3390/v6114731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Carbone A., Gloghini A., Dotti G. EBV-associated lymphoproliferative disorders: classification and treatment. Oncologist. 2008;13:577–585. doi: 10.1634/theoncologist.2008-0036. [DOI] [PubMed] [Google Scholar]
  • 91.Uldrick T.S., Whitby D. Update on KSHV-epidemiology, Kaposi sarcoma pathogenesis, and treatment of Kaposi sarcoma. Cancer Lett. 2011;305:150–162. doi: 10.1016/j.canlet.2011.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Szajerka T., Jablecki J. Kaposi’s sarcoma revisited. AIDS Rev. 2007;9:230–236. [PubMed] [Google Scholar]
  • 93••.Uldrick T.S., Polizzotto M.N., Aleman K., Wyvill K.M., Marshall V., Whitby D., Wang V., Pittaluga S., O’Mahony D., Steinberg S.M. Rituximab plus liposomal doxorubicin in HIV-infected patients with KSHV-associated multicentric Castleman disease. Blood. 2014;124:3544–3552. doi: 10.1182/blood-2014-07-586800. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates a strong clinical treatment for MCD with a combination therapy of Rituximab and doxorubicin.
  • 94.Diamantopoulos P.T., Polonyfi K., Sofotasiou M., Papadopoulou V., Kalala F., Iliakis T., Zervakis K., Tsilimidos G., Kouzis P., Kyrtsonis M.C. Rituximab in the treatment of EBV-positive low grade B-cell lymphoma. Anticancer Res. 2013;33:5693–5698. [PubMed] [Google Scholar]
  • 95.Song S.N.J., Tomosugi N., Kawabata H., Ishikawa T., Nishikawa T., Yoshizaki K. Down-regulation of hepcidin resulting from long-term treatment with an anti-IL-6 receptor antibody (tocilizumab) improves anemia of inflammation in multicentric Castleman disease. Blood. 2010;116:3627–3634. doi: 10.1182/blood-2010-03-271791. [DOI] [PubMed] [Google Scholar]
  • 96.Williams S.C.P. First IL-6–blocking drug nears approval for rare blood disorder. Nat Med. 2013;19:1193. doi: 10.1038/nm1013-1193. [DOI] [PubMed] [Google Scholar]
  • 97.Cohen J.I. In: Vaccine Development for Epstein-Barr Virus BT – Human Herpesviruses. Kawaguchi Y., Mori Y., Kimura H., editors. Springer; Singapore: 2018. pp. 477–493. [Google Scholar]
  • 98•.Baiocchi R.A., Ward J.S., Carrodeguas L., Eisenbeis C.F., Peng R., Roychowdhury S., Vourganti S., Sekula T., O’Brien M., Moeschberger M. GM-CSF and IL-2 induce specific cellular immunity and provide protection against Epstein-Barr virus lymphoproliferative disorder. J Clin Invest. 2001;108:887–894. doi: 10.1172/JCI12932. [DOI] [PMC free article] [PubMed] [Google Scholar]; Report showing that combined GM-CSF and low-dose IL-2 therapy can prevent the immune deficiencies that lead to fatal EBV-LPD in the hu-PBL-SCID mouse depleted of murine NK cells, and they point to a critical role for several human cellular subsets in mediating this protective effect.
  • 99.Hartlage A.S., Liu T., Patton J.T., Garman S.L., Zhang X., Kurt H., Lozanski G., Lustberg M.E., Caligiuri M.A., Baiocchi R.A. The Epstein-Barr virus lytic protein BZLF1 as a candidate target antigen for vaccine development. Cancer Immunol Res. 2015;3:787–795. doi: 10.1158/2326-6066.CIR-14-0242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Andrei G., De Clercq E., Snoeck R. In: Viral DNA Polymerase Inhibitors BT – Viral Genome Replication. Raney K.D., Gotte M., Cameron C.E., editors. Springer; US: 2009. pp. 481–526. [Google Scholar]
  • 101.Jamieson A.T., Gentry G.A., Subak Sharpe J.H. Induction of both thymidine and deoxycytidine kinase activity by herpes viruses. J Gen Virol. 1974;24:465–480. doi: 10.1099/0022-1317-24-3-465. [DOI] [PubMed] [Google Scholar]
  • 102.Li C., Quenelle D.C., Prichard M.N., Drach J.C., Zemlicka J. Synthesis and antiviral activity of 6-deoxycyclopropavir, a new prodrug of cyclopropavir. Bioorg Med Chem. 2012;20:2669–2674. doi: 10.1016/j.bmc.2012.02.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Coen N., Duraffour S., Naesens L., Krecmerova M., Van den Oord J., Snoeck R., Andrei G. Evaluation of novel acyclic nucleoside phosphonates against human and animal gammaherpesviruses revealed an altered metabolism of cyclic prodrugs upon Epstein-Barr virus reactivation in P3HR-1 cells. J Virol. 2013;87:12422–12432. doi: 10.1128/JVI.02231-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Zhang Y.-J., Patel D., Nan Y., Fan S. Inhibition of primary effusion lymphoma engraftment in SCID mice by morpholino oligomers against early lytic genes of Kaposi’s sarcoma-associated herpesvirus. Antivir Ther. 2011;16:657–666. doi: 10.3851/IMP1810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Zhang Y.-J., Wang K.-Y., Stein D.A., Patel D., Watkins R., Moulton H.M., Iversen P.L., Matson D.O. Inhibition of replication and transcription activator and latency-associated nuclear antigen of Kaposi’s sarcoma-associated herpesvirus by morpholino oligomers. Antivir Res. 2007;73:12–23. doi: 10.1016/j.antiviral.2006.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Hahn A.S., Desrosiers R.C. Binding of the Kaposi’s sarcoma-associated herpesvirus to the ephrin binding surface of the EphA2 receptor and its inhibition by a small molecule. J Virol. 2014;88:8724–8734. doi: 10.1128/JVI.01392-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107•.Schumann S., Jackson B.R., Yule I., Whitehead S.K., Revill C., Foster R., Whitehouse A. Targeting the ATP-dependent formation of herpesvirus ribonucleoprotein particle assembly as an antiviral approach. Nat Microbiol. 2016;2:16201. doi: 10.1038/nmicrobiol.2016.201. [DOI] [PMC free article] [PubMed] [Google Scholar]; This report demonstrates potent small molecule inhibitors of the cellular protein UAP56 that effectively inhibit KSHV lytic replication by targeting the ATP-dependent formation of viral ribonucleoprotein particles.
  • 108.Baquero-Pérez B., Whitehouse A. Hsp70 isoforms are essential for the formation of Kaposi’s sarcoma-associated herpesvirus replication and transcription compartments. PLoS Pathog. 2015;11 doi: 10.1371/journal.ppat.1005274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109•.Hughes D.J., Wood J.J., Jackson B.R., Baquero-Pérez B., Whitehouse A. NEDDylation is essential for Kaposi’s sarcoma-associated herpesvirus latency and lytic reactivation and represents a novel anti-KSHV target. PLoS Pathog. 2015;11:1–26. doi: 10.1371/journal.ppat.1004771. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study identifies a NEDDylation inhibitor as a novel KSHV theraputic by preventing the recruitment of the viral pre-replication complex to the origin of lytic DNA replication.
  • 110.Nichols L.A., Adang L.A., Kedes D.H. Rapamycin blocks production of KSHV/HHV8: insights into the anti-tumor activity of an immunosuppressant drug. PLoS One. 2011;6 doi: 10.1371/journal.pone.0014535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Chen J., Jiang L., Lan K., Chen X. Celecoxib inhibits the lytic activation of Kaposi’s sarcoma-associated herpesvirus through down-regulation of RTA expression by inhibiting the activation of p38 MAPK. Viruses. 2015;7:2268–2287. doi: 10.3390/v7052268. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES