Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Apr 30.
Published in final edited form as: Curr Opin Virol. 2022 Sep 27;56:101260. doi: 10.1016/j.coviro.2022.101260

Therapeutic Approaches to Epstein-Barr Virus Cancers

Samantha S Soldan 1, Troy E Messick 1, Paul M Lieberman 1
PMCID: PMC11058316  NIHMSID: NIHMS1985220  PMID: 36174496

Summary

Epstein–Barr virus (EBV) establishes a lifelong latent infection that can be a causal agent for a diverse spectrum of cancers and autoimmune disease. A complex and dynamic viral lifecycle evades eradication by the host immune system and confounds antiviral therapeutic strategies. To date, there are no clinically approved vaccines or therapies that selectively target EBV as the underlying cause of EBV-associated disease. Here, we review the challenges and recent advances in the development of EBV-specific therapeutics for treatment of EBV-associated cancers.

Epstein–Barr virus prevalence, lifecycle, and disease burden

Epstein–Barr virus (EBV) is a human gamma-herpesvirus that establishes lifelong latency in over 95% of the adult population [1]. EBV is also an etiological agent for ~2% of all human cancers, most notably endemic Burkitt’s lymphoma, 50% of Hodgkin’s lymphoma (HL), ~10% of gastric carcinomas (GC), and undifferentiated nasopharyngeal carcinomas (NPC) [2,3]. In addition, EBV is a triggering factor in autoimmune disorders, especially multiple sclerosis (MS) [4,5]. EBV has a complex lifecycle that enables high-transmissibility and lifelong persistence [1]. These attributes, combined with geographical, socioeconomic, and genetic variations in virus and host, contribute to the challenges of developing successful and comprehensive EBV-selective therapeutics.

The complex EBV lifecycle presents many challenges to therapeutic strategies. After primary infection in the oropharynx, the virus enters and replicates in oropharyngeal epithelial cells and infects local infiltrating B-lymphocytes. EBV infection of resting B-cells through CD21 receptor leads to a germinal center-like hyperproliferation followed by the emergence of slower cycling and long-lived memory B-cells [1,6]. During latency, EBV can adopt different gene-expression programs that are associated with the different infected cell and tumor types [7]. EBV latent genes drive B-cell proliferation and persistence in reservoirs of long-lived memory B-cells that evade host immune elimination. Viral reactivation can occur through several pathways, including the terminal differentiation of latently infected memory B-cells. Numerous viral genes have been implicated in EBV-disease pathogenesis, including those associated with latent and lytic cycle, and it remains unclear which viral genes provide the best targets for therapeutic intervention [8,9]. Here, we review recent efforts in vaccines, cellular–immune-based therapies, and small molecules to treat EBV-associated cancers.

Epstein–Barr virus therapeutics

Vaccines

Several vaccine strategies to prevent EBV infection or treat EBV-associated disease are under investigation (Table 1). While vaccines to human herpesvirus 3 (Varicella-Zoster virus (VZV)) have demonstrated remarkable efficacy in preventing disease from primary childhood infection and late-life reactivation, this success has not yet been repeated for other members of the herpesvirus family [1012]. Attenuated live virus has been used successfully as the basis for other herpes-viruses vaccines, including Marek’s disease virus and VZV, but this strategy is technically and clinically challenging for EBV because of the relatively low yield and potential residual oncogenic activity of an attenuated virus. Consequently, better-defined viral-like particles (VLPs) and non-EBV viral vectors have received more attention for EBV vaccines. EBV-derived VLPs, including EBV mutants with deletions in oncogenic genes or DNA-packaging genes [13], have been produced by inducing cell lines to enter the lytic phase and subsequently purifying VLPs from cell supernatants. Several of these EBV VLPs also contain deletions in key lytic genes (BFLF1/BFRF1, BBRF1, BFLF2, and terminal repeats) to prevent virus replication and packaging to address safety concerns [13]. Recently, more immunogenic EBV VLPs were generated by fusing latent antigens (e.g. Epstein-Barr Nuclear Antigen 1 (EBNA1) and EBNA3C) to the tegument protein BNRF1 to stimulate a greater CD4+ T-cell response [14]. As an alternative to EBV-based VLPs, a New Castle disease virus VLP platform to generate an EBV-specific immune response was developed. NDP–VLP-based vaccines, including the EBV membrane antigen BLLF1 (gp350) ectodomain in combination with additional envelope proteins and latent antigens (e.g. gH/gL-EBNA1 and gB-LMP2), generate high titers of neutralizing antibodies and EBV-specific T-cell responses in mouse models, representing a safe and rapid method of producing VLPs for EBV vaccines [15].

Table 1.

Selection of completed and ongoing EBV vaccine trials

NIH Clinical Trials Identifier Vaccine platform EBV antigen Dose and route application Outcome measures Results Status as of January, 2022
NCT04645147 Ferritin nanoparticle gp350 3 doses, i.m. IM Ab and CD4+ to gp350 pending Recruiting
NCT05164094 mRNA gH, gL, gp220 3 doses, i.m. IM Immune response pending Recruiting
NCT00430534 recombinant gp350 3 doses, i.m. IM Immune response, development of IM, seroconversion prevents IM, does not prevent asymptomatic infection Completed (Phase II; Sokal, 2007)
NCT01094405 MVA EBNA1/LMP1 3 doses, i.d. NPC Stable disease, immune response, survival Increased CD4+, CD8+, and Ab Completed (Phase I; Hui 2013))

Recombinant protein vaccination studies have focused mostly on gp350 to prevent EBV infection. Early studies showed that purified gp350 elicited sufficient immunity to protect against EBV-induced malignant lymphomas [16]. Further work demonstrated that a multivalent, tetrameric gp350 created by fusing two gp350 proteins to a C-terminal leucine zipper with or without specific T-cell epitopes, enhanced neutralizing antibody responses in rabbits [17]. In addition, gp350 nanoparticles that induce high neutralizing antibody titers and provide protection against EBV infection in cynomolgus macaques have been produced [18]. Other studies have used other EBV glycoproteins and latent proteins, including gH/gL and gp42, as immunogens to elicit high neutralizing antibody titers. Ferritin-based nanoparticles containing fusion-apparatus component gH/gL or gH/ gL/gp42 were found to provide protection against EBV infection of B-cells and epithelial cells in culture [19]; a clinical trial with ferritin–gp350 is underway (Table 1) [10].

Most recently, nucleic-acid-based vaccines have come to the fore with the rapid development, success, and widespread use of mRNA vaccines for SARS-CoV-2 [20,21]. DNA vaccines using three EBV-latency genes (EBNA1, Latent membrane protein 1 (LMP1), and LMP2A) or a combined EBV–LMP2A–CD40L plasmid have been tested [22,23]. While LMP1 vaccination was antigenically weak and did not provide robust T-cell immunity, EBNA1 and LMP2A DNA-based vaccines were highly immunogenic, induced dominant CD8+ T-cell responses, and prevented tumor growth [22,24]. Based on the recent success of mRNA vaccines for SARS-Cov2, an EBV-targeted mRNA vaccine encoding the major EBV glycoproteins (gp350, gB, gH/gL, and gp42) is being tested for safety and reactogenicity among healthy participants ages of 18–30 (Table 1).

Cellular–immune-therapy approaches

Cellular immunotherapies are in development for the treatment of EBV-associated cancers and autoimmune disorders. Cellular–immune therapy may be achieved by delivering antigen-presenting cells loaded with EBV tumor antigens or by vaccination. Alternatively, cytotoxic cells (cytotoxic T-cell lines (CTLs) or natural killer (NK) cells), from adoptive transfer of the patient’s cells or from an Human Leukocyte Antigens (HLA)-matched donor that are primed and expanded to target and kill cancer cells or, in the case of multiple sclerosis, latently infected EBV-positive B-cells, are also under investigation [25,26]. Finally, EBV-specific allogeneic T-cells/ chimeric antigen-receptor T-cells (CAR T-cells) genetically engineered to produce an artificial TCR for use in immunotherapy are under development with EBV antigen (EBNA3C, LMP1, and gp350)-specific chimeric antigen receptors and showed good efficacy in preclinical models [2730] (Table 2).

Table 2.

Selection of cell-based immunotherapy trials for EBV-associated diseases

NIH Clinical Trials Identifier Intervention Platform Dose and route application Outcome measures Results Status as of January, 2022
NCT03648697 Adoptive T-cell immunotherapy LMP1, LMP2, and EBNA1-TCR-T cells 1 dose, i.v. NPC Adverse events, clinical pending Recruiting
NCT03925896 Adoptive T-cell immunotherapy LMP2 specific TCR-T cells 1 dose, i.v. NPC Maximum Tolerated Dose pending Recruiting
NCT00430534 Adoptive T-cell immunotherapy gemcitabine-carboplatin followed by EBV-specific CTL Multiple doses gemcitabine-carboplatin, Followed by 4 doses of EBV-specific CTL NPC Survival, disease progression pending Active (Phase III)
NCT01498484 Adoptive T-cell immunotherapy EBV specific CTLs from HLA matched donor (ATA129) 3 doses, i.v. lymphoma Clinical response rate Complete or partial remission in over 58% Completed (Phase II; Prockop, 2020)
NCT03283826 Adoptive T-cell immunotherapy EBV specific CTLs from HLA matched donor (ATA129) 3 doses, i.v. MS Adverse events, clinical progression (EDSS), MRI, IgG Index pending Recruiting

Donor-derived EBV-specific CTLs have proven successful in the treatment of post-transplantation lymphoproliferative disorders, with low rates of graft-versus-host disease [3133]. In addition, the adoptive transfer of CTLs has been used for various EBV-associated cancers and, especially lymphomas [34]. In these studies, a more restricted group of EBV antigens is expressed and CTLs specific for EBNA1, LMP1, and LMP2 have shown clinical efficacy in EBV-associated lymphomas and NPC [3537]. For patients with advanced EBV-positive NPC, EBV-specific CTLs were administered in combination with chemotherapy (gemcitabine and carboplatin) with promising response and survival rates and some patients experiencing stable disease and reduced tumor growth [38]. In another Phase-I/II clinical trial, EBV-specific CTL therapy, generated using the patients’ own EBV (B95–8)-transformed LCLs to generate and expand CTLs, was used alone in 21 patients with recurrent, metastatic NPC [39]. Although the overall response rate was low, a subset of patients demonstrated robust responses to chemotherapy regimens that they had previously failed, suggesting that the enhancement of the immune response by EBV-specific CTLs had a broad effect that restored the patients’ response to conventional chemotherapy [39].

Checkpoint inhibitors

Most cancer types, including EBV cancers, are known to upregulate immune-checkpoint ligands, especially PD-L1, making them susceptible to immune-checkpoint blockade, either alone or in combination with other antiviral treatment modalities [4043]. Immune-checkpoint therapy is under investigation for the treatment of EBVaGC [44]. PD-L1 expression is frequently observed in EBVaGC by immunohistochemistry [45,46] and EBVaGC is responsive to the antiPD-L1 antibody avelumab [47]. In addition, a Phase-II trial of pembrolizumab (antiPD-1 antibody) monotherapy for metastatic gastric carcinoma demonstrated that a EBVaGC was more responsive (100%) than EBV-negative gastric cancers (50–85.7%).

Based on the prognostic value of PD-L1 in NPC, the PD-1/PD-L1 checkpoint has been extensively investigated in NPC. More than 70% of NPC patients present with advanced disease at diagnosis and chemoradiotherapy has limited efficacy for advanced metastatic disease [48]. Higher levels of tumor-infiltrating lymphocytes have been described in EBV-positive NPC compared with EBV-negative NPCs. Accordingly, CD8+ cells within EBV-positive NPC tumors are associated with higher expression of PD-L1 (found in 50–80% of NPC tissues and preclinical models), CD68+ tumor-associated macrophages and Forkhead box P-3+ Tregs, T-cell immunoglobulin mucin-3, and lymphocyte-activating 3 [49] and CD4+ cells with CTLA4 [50]. Combinatorial approaches using antibodies targeting the PD-1/PD-L1 checkpoint with chemotherapeutic agents, including cisplatin, may improve outcomes by enhancing tumor recognition and further decreasing the immunosuppressive milieu of the tumor microenvironment [51,52]. Clinical trials determining the combined inhibition of PD-1 with CTLA4, DKY709 (an immunomodulatory agent that targets Tregs), and Tabelecleucel (an EBV-specific CTL therapy generated from healthy donors) are underway [41].

Immune-checkpoint inhibitors are also of interest for their therapeutic potential in EBV-associated lymphomas. EBV LMP1 and LMP2 upregulate PD-L1 expression in a subset of classic HL patients, making these lymphomas sensitive to PD-1 blockade [53]. HL patients have been demonstrated to be responsive to nivolumab (PD-1 inhibitor) and pembrolizumab (PD-L1 inhibitor) [54,55] and, consequently, both drugs were given accelerated FDA approval for the treatment of relapsed/refractory HL. However, PD-L1 is upregulated in both EBV-negative and EBV-positive tumors, as well in the surrounding lymphocytes [56]. Therefore, it has yet to be determined whether the high sensitivity of EBV-positive HL to nivolumab and pembrolizumab is specifically counteracting PD-L1 induction by EBV LMP1/2 in these tumors. High RNA expression of PD-1 and PD-L2 has been demonstrated in Primary Central Nervous System Lymphoma (PCNSL) brain specimens and this high expression is associated with a poor prognosis [57]. AntiPD-1 antibodies were tested in a pre-clinical model of CNS lymphoma using murine lymphoma cells that express PD-L1 [58]. Although immune-checkpoint inhibitors have not been rigorously investigated in EBV+ PCNSL, anecdotal reports have shown promise and the available data support the further development of PD-1/PD-L1 inhibitors against EBV-positive PCNSL in the CNS [59].

Small-molecule approaches (viral targets)

Small molecules have been highly successful for targeted treatment of a few viruses, notably Human Immunodeficiency Virus (HIV), Hepatitis C virus (HCV), Herpes Simplex Virus (HSV), and most recently SARS-CoV-2. To date, there are no FDA-approved small molecules that selectively target EBV and those used to treat EBV-associated cancers have no known selectivity for the virus. Most EBV-associated disease is treated agnostically with regard to EBV status. However, EBV-positive tumors provide several unique viral targets and there are several EBV-specific treatment strategies under investigation with some innovative therapeutic approaches entering clinical trials (Table 3).

Table 3.

Selection of clinical trials for small molecule approaches to EBV-associated malignancies

NIH Clinical Trials Identifier Compound Class application Status as of January, 2022
NCT05011058 Nanatinostat Class I HDAC inhibitor In combination with Valganciclovir in patients with relapsed/refractory EBV-positive lymphomas, including: EBV- associated lymphoproliferative disease; PTLD, Non-Hodgkin lymphoma, extranodal NK/T, DLBC, Hodgkin lymphoma, PTCL Recruiting (Phase II)
NCT04925544 VK-2019 EBNA1 inhibitor Advanced NPC Recruiting (Phase II)
NCT02670616 Ibrutinib BTK inhibitor In combination with Rituximab-CHOP for diffuse large B-cell lymphoma Completed (Phase II)
NCT01094405 HQK-1004 HDAC inhibitor In combination with Valganciclovir for relapsed/refractory EBV lymphoid malignancies and lymphoproliferative disorders Completed (Phase II)

References Sokal EM, Hoppenbrouwers K, Vandermeulen C, Moutschen M, Léonard P, Moreels A, Haumont M, Bollen A, Smets F, Denis M. Recombinant gp350 vaccine for infectious mononucleosis: a phase 2, randomized, double-blind, placebo-controlled trial to evaluate the safety, immunogenicity, and efficacy of an Epstein-Barr virus vaccine in healthy young adults. J Infect Dis. 2007 Dec 15;196(12):1749–53. doi: 10.1086/523813.

Edwin P. Hui, Graham S. Taylor, Hui Jia, Brigette B.Y. Ma, Stephen L. Chan, Rosalie Ho, Wai-Lap Wong, Steven Wilson, Benjamin F. Johnson, Ceri Edwards, Deborah D. Stocken, Alan B. Rickinson, Neil M. Steven and Anthony T.C. Chan Phase I Trial of Recombinant Modified Vaccinia Ankara Encoding Epstein–Barr Viral Tumor Antigens in Nasopharyngeal Carcinoma Patients. Cancer Res. 2013. PMID: 23348421

Prockop S, Doubrovina E, Suser S, Heller G, Barker J, Dahi P, Perales MA, Papadopoulos E, Sauter C, Castro-Malaspina H, Boulad F, Curran KJ, Giralt S, Gyurkocza B, Hsu KC, Jakubowski A, Hanash AM, Kernan NA, Kobos R, Koehne G, Landau H, Ponce D, Spitzer B, Young JW, Behr G, Dunphy M, Haque S, Teruya-Feldstein J, Arcila M, Moung C, Hsu S, Hasan A, O’Reilly RJ. Off-the-shelf EBV-specific T cell immunotherapy for rituximab-refractory EBV-associated lymphoma following transplantation. J Clin Invest. 2020 Feb 3;130(2):733–747. doi: 10.1172/JCI121127.

BXLF1 and BGLF4 (viral-encoded kinases)

EBV-positive tumor cells express a limited number of EBV-encoded latent genes with relatively few tumor cells expressing lytic genes. One approach is to induce the lytic cycle and viral proteins that are susceptible to small-molecule inhibition [60]. This ‘kick and kill’ strategy pushes the virus into lytic replication, rendering EBV vulnerable to inhibitors of lytic enzymes, especially viral kinases and DNA polymerases [6164]. For EBV cancers, incorporation of nucleoside-chain terminators has the added benefit of inhibiting cellular DNA replication, providing viral-specific antitumor activity.

Histone deacetylase (HDAC) inhibitors are known to be potent activators of the lytic cycle [65]. Lytic replication is dependent on the expression of two immediate–early genes — BZLF1 and BRLF1. HDAC inhibitors derepress the promoters of these two genes, allowing the expression of Zta and Rta to activate a cascade of lytic genes, resulting in the production of viral-encoded replication enzymes [6668]. During lytic induction, EBV expresses BamHI X Left Frame 1 (BXLF1) and BamHI G Left Frame 4 (BGLF4) that encode for thymidine kinase and protein kinase (PK). EBV-PK phosphorylates and converts the nucleoside analogs into their active, cytotoxic form in EBV-infected cells [69]. Aciclovir and ganciclovir and their prodrug forms valciclovir and valganciclovir are analogs of 2′-deoxyguanosine. After phosphorylation to the monophosphate form by EBV-PK, cellular kinases convert the analogs to the active triphosphate forms, which are incorporated by viral and cellular DNA polymerases into the replicating viral and cellular DNA, resulting in chain termination that preferentially blocks viral DNA replication, but can also cause host cell-cycle arrest and apoptosis [70]. Viral resistance to various PK inhibitors has been identified and represents a challenge for long-term treatments [70].

Several noncyclic nucleoside analogs, such as tenofovir alafenamide (TAF), that were developed as specific inhibitors of reverse transcriptases to treat HIV and HBV infection, were identified as potent inhibitors of EBV DNA polymerase [71]. TAF was found to be twice as potent as ganciclovir in direct inhibition of EBV DNA polymerase activity in vitro and viral DNA replication in cell culture [72]. There is some evidence suggesting that tenofovir may provide some benefit for treatment of MS and may be related to its antiviral activity directed toward lytic EBV [73].

An early clinical trial on the use of arginine butyrate in combination with ganciclovir showed modest therapeutic benefit for EBV lymphoma [63]. Further optimization of HDAC and nucleoside analog improved efficacy, especially in EBV-positive NK/T-cell lymphoma. Presently, a Phase-2 clinical trial is in progress for the HDAC inhibitor nanatinostat in combination with valganciclovir to determine its efficacy in various EBV-positive hematological malignancies, including PTLD, Diffuse Large B-Cell Lymphoma (DLBCL), and HL. Another HDAC inhibitor, HQK-1004, has also been tested in combination with valganciclovir in patients with relapsed or refractory EBV-positive lymphoid malignancies or other lymphoproliferative disorders. HDAC inhibitors have been found to induce cellular differentiation of EBV-positive NPC, suggesting they may provide direct antineoplastic activity independent of viral reactivation [74].

In addition to HDACs, numerous other agents have been found to induce the EBV lytic cycle with some dependence on host-cell type. A class of thiosemicarbazone that chelate intracellular iron was found to be potent activator of EBV and has been explored therapeutically [75,76]. A family of tetrahydro-β-carbolines, with a lead termed C60, were found to induce EBV lytic reactivation through protein stabilization of BZLF1 (ZTA) and a mechanism involving perturbation of Cullin-associated and neddylation-dissociated 1 protein (CAND1)-dependent regulation of CRL F-box ubiquitin-ligation complex [7779].

There are some limitations to this approach. Response to lytic induction is heterogeneous with on average 2–60% of tumor cells expressing lytic genes, depending on which lytic activator drug is used. Lytic activation is also variable in terms of the cellular context with some cell lines (many derived from solid tumors) only weakly induced [67,80,81]. Common side effects from the HDAC inhibitor, vorinostat Suberoylanilidine hydroxamic acid (SAHA), for example, include anemia, thrombocytopenia, and intestinal issues [82]. It is also not yet known if the systemic reactivation of EBV may increase risk for subsequent malignancies or immune disorders.

EBNA1 (DNA-binding episome maintenance)

EBNA1, a viral-encoded regulatory protein critical for the replication, maintenance, and survival of EBV during latency. EBNA1 is the only protein that is expressed in all oncogenic forms of latency and in all EBV-positive tumors [83]. Thus, EBNA1 may be considered an attractive target for therapeutic intervention. The structure of the C-terminal domain of EBNA1 has been solved alone and bound to cognate DNA [8486]. The C-terminal DNA-binding domain of EBNA1 is an obligate homodimer. Small molecules or peptides that perturb the dimer–dimer interface have activity in vitro and in vivo [8789]. Several pockets in the dimeric C-terminal domain were found to be susceptible to small-molecule binding and inhibition of DNA binding [90,91]. Using a combination of fragment-based lead discovery and structure-based design, a unique series of 2-,3-substituted benzoic acids was found to inhibit EBNA1–DNA binding [92]. These inhibitors blocked EBNA1–DNA interaction in vitro and EBNA1-dependent replication and oriP binding in cellular assays. Moreover, EBNA1 inhibitors specifically blocked the proliferation of EBV-positive cells in cellular assays and preclinical models [92,93]. Treatment with EBNA1 inhibitors also resulted in significant loss of EBV genomes and viral gene expression. One EBNA1 inhibitor (VK-2019) has progressed to clinical studies for treatment of patients with advanced NPC (Table 3).

BILF1 (viral G-protein-coupled receptor)

EBV encodes a G-protein-coupled receptor (GPCR), BamHI I Left Frame 1 (BILF1), that can be expressed at variable levels in different infection scenarios and tumor types. BILF1 is a lytic protein that downregulates the expression of a broad range of surface HLA class-I molecules and impedes presentation of viral antigens, allowing the virus to evade cytotoxic T-cells [94,95]. GPCRs are 7-transmembrane proteins that are frequently targeted by small-molecule inhibitors. Small molecules that bind and inhibit BILF1 signaling are under development to treat EBV-driven disease [96].

Latent membrane protein1/LMP2A (viral membrane oncogenes)

Latent membrane protein 1 (LMP1) is a functional homolog and acts as a constitutively active receptor of CD40, recruiting cellular signaling molecules associated with tumor necrosis factor receptors. Together with LMP2A, LMP1 activates numerous pathways, including Nuclear Factor-kappa B (NF-κB), phosphatidylinositol-3-kinase, mitogen-activated protein kinase, Interferon Regulated Factor 7 (IRF7), and Signal Transducers and Activators of Transcription (STAT), and drives cellular survival and proliferation. Recently, affibody molecules that interact with LMP2A N-terminal or C-terminal domains were shown to inhibit proliferation of NPC cells [97,98].

Other Epstein–Barr virus-encoded targets

EBV encodes many additional proteins and noncoding RNAs that are implicated in cancer pathogenesis that represent attractive targets for small-molecule inhibition. These include BamHI A Right Frame 1 (BARF1), a Cytokine Stimulatory Factor 1 (CSF1)-interacting protein expressed in many EBV-epithelial tumors [99], latency-associated nuclear regulatory protein Epstein-Barr Nuclear Antigen 2 (EBNA2) [100], the viral-encoded ubiquitin ligase BamHI P Left Frame 1 (BPLF1) [101], and the viral-encoded ribonucleotide reductase BamHI O Right Frame 2 (BORF2) that also inhibits APO-BEC3B [102], to name just a few. It is not yet clear which of these early-stage targets are most likely to produce an efficacious inhibitor to treat EBV-driven cancers.

Conclusions

Selective therapies to treat EBV-associated disease have been challenged by the complexity of the EBV lifecycle, host immunity, the heterogeneity of viral gene expression, and the diversity of diseases caused by EBV infection. Various immune strategies are likely to be effective in reducing transmission and disease burden and provide new and safe methods to treat EBV malignancies. Small molecules targeting EBV may also provide selective modalities that, in combination with existing cancer therapies, or with newly developed immune approaches, may provide precision approaches for EBV-driven cancers and autoimmune disorders.

Acknowledgements

The authors have received funding from the following grants: National Institute of Health, USA (NIH) DE017336, AI153508, CA140652, CA093606, CA259171, and Commonwealth of Pennsylvania — CURE Formula Funding SAP# 4100088567 to PML.

Footnotes

Conflict of interest statement

The authors PML and TEM hold patents on EBNA1 inhibitors that are in clinical trial with potential for royalty payments. PML is a founder and advisor of Vironika, LLC.

References and recommended reading

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

• of special interest

•• of outstanding interest.

  • 1.Thorley-Lawson DA: EBV persistence–introducing the virus. Curr Top Microbiol Immunol 2015, 390:151–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Shannon-Lowe C, Rickinson A: The global landscape of EBV-associated tumors. Front Oncol 2019, 9:713–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Wong Y, Meehan MT, Burrows SR, Doolan DL, Miles JJ: Estimating the global burden of Epstein-Barr virus-related cancers. J Cancer Res Clin Oncol 2022, 148:31–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bjornevik K, Cortese M, Healy BC, Kuhle J, Mina MJ, Leng Y, Elledge SJ, Niebuhr DW, Scher AI, Munger KL, et al. : Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science 2022, 375:296–301. [DOI] [PubMed] [Google Scholar]
  • 5.Ruprecht K: The role of Epstein-Barr virus in the etiology of multiple sclerosis: a current review. Expert Rev Clin Immunol 2020, 16:1143–1157. [DOI] [PubMed] [Google Scholar]
  • 6.Thorley-Lawson DA: Epstein-Barr virus: exploiting the immune system. Nat Rev Immunol 2001, 1:75–82. [DOI] [PubMed] [Google Scholar]
  • 7.Kanda T: EBV-encoded latent genes. Adv Exp Med Biol 2018, 1045:377–394. [DOI] [PubMed] [Google Scholar]
  • 8.Kieff E, Rickinson AB: Epstein-Barr virus and its replication. In Fields Virology. Edited by Knipe DM, Howley PM. Lippincott Williams and Wilkins; 2007:2603–2654. [Google Scholar]
  • 9.Farrell PJ: Epstein-Barr virus and cancer. Annu Rev Pathol 2019, 14:29–53. [DOI] [PubMed] [Google Scholar]
  • 10.Cohen JI: Vaccine development for Epstein-Barr virus. Adv Exp Med Biol 2018, 1045:477–493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Willis ED, Woodward M, Brown E, Popmihajlov Z, Saddier P, Annunziato PW, Halsey NA, Gershon AA: Herpes zoster vaccine live: a 10year review of post-marketing safety experience. Vaccine 2017, 35:7231–7239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Warren-Gash C, Forbes H, Breuer J: Varicella and herpes zoster vaccine development: lessons learned. Expert Rev Vaccin 2017, 16:1191–1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sun C, Chen XC, Kang YF, Zeng MS: The status and prospects of Epstein-Barr virus prophylactic vaccine development. Front Immunol 2021, 12:677027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • ••14.van Zyl DG, Tsai MH, Shumilov A, Schneidt V, Poirey R, •• Schlehe B, Fluhr H, Mautner J, Delecluse HJ: Immunogenic particles with a broad antigenic spectrum stimulate cytolytic T cells and offer increased protection against EBV infection ex vivo and in mice. PLoS Pathog 2018, 14:e1007464. [DOI] [PMC free article] [PubMed] [Google Scholar]; Using a comprehensive strategy that primes the immune system against both lytic and latent EBV antigens, the authors describes the generation of immunogenic virus-like particles that contain latent (envelope, tegument, and capsid) and latency proteins (EBNA1 and EBNA3C) These virus-like particles enabled ex vivo expansion of EBV-specific T cells that efficiently recognize and control EBV-infected B cells in vitro. Notably, these VLPs were effective against wild-type EBV infection in humanized mice. This study underscores the importance of include proteins spanning the EBV lifecycle in vaccine design.
  • 15.Freer G, Pistello M: Varicella-zoster virus infection: natural history, clinical manifestations, immunity and current and future vaccination strategies. New Microbiol 2018, 41:95–105. [PubMed] [Google Scholar]
  • 16.Finerty S, Tarlton J, Mackett M, Conway M, Arrand JR, Watkins PE, Morgan AJ: Protective immunization against Epstein-Barr virus-induced disease in cottontop tamarins using the virus envelope glycoprotein gp340 produced from a bovine papillomavirus expression vector. J Gen Virol 1992, 73:449–453. [DOI] [PubMed] [Google Scholar]
  • •17.Cui X, Cao Z, Chen Q, Arjunaraja S, Snow AL, Snapper CM: • Rabbits immunized with Epstein-Barr virus gH/gL or gB recombinant proteins elicit higher serum virus neutralizing activity than gp350. Vaccine 2016, 34:4050–4055. [DOI] [PubMed] [Google Scholar]; Although many early vaccine studies focused solely on the EBV membrane antigen BLLF1 (gp350) ectodomain, monomeric p350 vaccines have had modest effect in clinical trials. This study used a rabbit model of EBV infection to demonstrate the increased immunogeneicity of trimeric and monomeric gH/gL, trimeric gB, and tetrameric gp350 compared to monomeric gp350. These studies present a strong case for including gH/gL, and gB as components of EBV vaccine strategies.
  • 18.Kanekiyo M, Bu W, Joyce MG, Meng G, Whittle JR, Baxa U, Yamamoto T, Narpala S, Todd JP, Rao SS, et al. : Rational design of an Epstein-Barr virus vaccine targeting the receptor-binding site. Cell 2015, 162:1090–1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • ••19.Bu W, Joyce MG, Nguyen H, Banh DV, Aguilar F, Tariq Z, Yap ML, •• Tsujimura Y, Gillespie RA, Tsybovsky Y, et al. : Immunization with components of the viral fusion apparatus elicits antibodies that neutralize Epstein-Barr virus in B cells and epithelial cells. Immunity 2019, 50:1305–1316 e1306. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper shows that an EBV nanoparticle vaccine to viral fusion apparatus (gH/gL or gH/gL/gp42) elicit antibodies that inhibit virus-fusion and infection broadly across multiple cell types, and in contrast to EBV gp350 vaccine candidates, which only block infection in B cells.
  • 20.Polack FP, Thomas SJ, Kitchin N, Absalon J, Gurtman A, Lockhart S, Perez JL, Perez Marc G, Moreira ED, Zerbini C, et al. : Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med 2020, 383:2603–2615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Jackson LA, Anderson EJ, Rouphael NG, Roberts PC, Makhene M, Coler RN, McCullough MP, Chappell JD, Denison MR, Stevens LJ, et al. : An mRNA vaccine against SARS-CoV-2-preliminary report. N Engl J Med 2020, 383:1920–1931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •22.Wojtak K, Perales-Puchalt A, Weiner DB: Novel synthetic DNA • immunogens targeting latent expressed antigens of Epstein-Barr virus elicit potent cellular responses and inhibit tumor growth. Vaccines 2019, 7:44–60. [DOI] [PMC free article] [PubMed] [Google Scholar]; Using novel DNA vaccine technology, this paper tests the immunogenicity of DNA vaccines against EBV EBNA1, LMP1 and LMP2 delivered by electroporation. EBNA1VAX and LMP2aVAX were generated a superior T cell response compared to LMP1VAX. Moreover, B57/BL6 mice receiving three biweekly vaccinations of LMP2AVAX prior to engraftment of TC-1-LMP2A cells (derived from the epithelium of C57/BL6 mice and transduced to express LMP2A) successfully eliminated tumor cells.
  • 23.Li W, Chen Q, Chen H, Rao P, Xue X, Chen S, Zhu S, Zhang L: Immune response of mice to a latency membrane protein 2 multiepitope antigen of Epstein-Barr virus applied as DNA vaccine and/or peptide vaccine. Acta Virol 2013, 57:51–58. [DOI] [PubMed] [Google Scholar]
  • 24.Lei L, Li J, Liu M, Hu X, Zhou Y, Yang S: CD40L-adjuvanted DNA vaccine carrying EBV-LMP2 antigen enhances anti-tumor effect in NPC transplantation tumor animal. Cent Eur J Immunol 2018, 43:117–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Comoli P, De Palma R, Siena S, Nocera A, Basso S, Del Galdo F, Schiavo R, Carminati O, Tagliamacco A, Abbate GF, et al. : Adoptive transfer of allogeneic Epstein-Barr virus (EBV)-specific cytotoxic T cells with in vitro antitumor activity boosts LMP2-specific immune response in a patient with EBV-related nasopharyngeal carcinoma. Ann Oncol 2004, 15:113–117. [DOI] [PubMed] [Google Scholar]
  • 26.Comoli P, Pedrazzoli P, Maccario R, Basso S, Carminati O, Labirio M, Schiavo R, Secondino S, Frasson C, Perotti C, et al. : Cell therapy of stage IV nasopharyngeal carcinoma with autologous Epstein-Barr virus-targeted cytotoxic T lymphocytes. J Clin Oncol 2005, 23:8942–8949. [DOI] [PubMed] [Google Scholar]
  • •27.Chicaybam L, Abdo L, Carneiro M, Peixoto B, Viegas M, • de Sousa P, Fornazin MC, Spago MC, Albertoni Laranjeira AB, de Campos-Lima PO, et al. : CAR T cells generated using sleeping beauty transposon vectors and expanded with an EBV-transformed lymphoblastoid cell line display antitumor activity in vitro and in vivo. Hum Gene Ther 2019, 30:511–522. [DOI] [PubMed] [Google Scholar]; This study shows the use of EBV+ LCLs as the basis for CAR T cell technology. Here, LCLs were used to develop 19BBζ CAR T cells that demonstrated high cytotoxic activity against CD19+ cells. These cells showed high rates of proliferation and survival and conferred protection against a human preB leukemic cell line (RS4;11) in NSG mice.
  • 28.McLaughlin LP, Rouce R, Gottschalk S, Torrano V, Carrum G, Wu MF, Hoq F, Grilley B, Marcogliese AM, Hanley PJ, et al. : EBV/ LMP-specific T cells maintain remissions of T- and B-cell EBV lymphomas after allogeneic bone marrow transplantation. Blood 2018, 132:2351–2361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Slabik C, Kalbarczyk M, Danisch S, Zeidler R, Klawonn F, Volk V, Kronke N, Feuerhake F, Ferreira de Figueiredo C, Blasczyk R, et al. : CAR-T cells targeting Epstein-Barr virus gp350 validated in a humanized mouse model of EBV infection and lymphoproliferative disease. Mol Ther Oncolytics 2020, 18:504–524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • ••30.Dragon AC, Zimmermann K, Nerreter T, Sandfort D, Lahrberg J, •• Kloss S, Kloth C, Mangare C, Bonifacius A, Tischer-Zimmermann S, et al. : CAR-T cells and TRUCKs that recognize an EBNA-3C-derived epitope presented on HLA-B*35 control Epstein-Barr virus-associated lymphoproliferation. J Immunother Cancer 2020, 8:e000736. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study describes a novel peptide-selective chimeric antigen receptor (CAR) based on the monoclonal antibody TÜ165, which recognizes EBNA3C/HLA complexes (HLA-B35*) in a TCR-like manner. TÜ165 CARTs with inducible IL-12 expression were found to further enhance responses by recruiting additional immune cells. This strategy is especially promising for EBV associated post-transplant lymphoproliferative disease because it is designed to selectively targeting EBV+ cells while recruiting immune cells to limit EBV-driven lymphoproliferation.
  • 31.Doubrovina E, Oflaz-Sozmen B, Prockop SE, Kernan NA, Abramson S, Teruya-Feldstein J, Hedvat C, Chou JF, Heller G, Barker JN, et al. : Adoptive immunotherapy with unselected or EBV-specific T cells for biopsy-proven EBV+ lymphomas after allogeneic hematopoietic cell transplantation. Blood 2012, 119:2644–2656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Heslop HE, Slobod KS, Pule MA, Hale GA, Rousseau A, Smith CA, Bollard CM, Liu H, Wu MF, Rochester RJ, et al. : Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood 2010, 115:925–935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Savoldo B, Goss JA, Hammer MM, Zhang L, Lopez T, Gee AP, Lin YF, Quiros-Tejeira RE, Reinke P, Schubert S, et al. : Treatment of solid organ transplant recipients with autologous Epstein Barr virus-specific cytotoxic T lymphocytes (CTLs). Blood 2006, 108:2942–2949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Gottschalk S, Rooney CM: Adoptive T-Cell immunotherapy. Curr Top Microbiol Immunol 2015, 391:427–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Icheva V, Kayser S, Wolff D, Tuve S, Kyzirakos C, Bethge W, Greil J, Albert MH, Schwinger W, Nathrath M, et al. : Adoptive transfer of epstein-barr virus (EBV) nuclear antigen 1-specific t cells as treatment for EBV reactivation and lymphoproliferative disorders after allogeneic stem-cell transplantation. J Clin Oncol 2013, 31:39–48. [DOI] [PubMed] [Google Scholar]
  • 36.Bollard CM, Tripic T, Cruz CR, Dotti G, Gottschalk S, Torrano V, Dakhova O, Carrum G, Ramos CA, Liu H, et al. : Tumor-specific T-cells engineered to overcome tumor immune evasion induce clinical responses in patients with relapsed Hodgkin lymphoma. J Clin Oncol 2018, 36:1128–1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Smith C, Tsang J, Beagley L, Chua D, Lee V, Li V, Moss DJ, Coman W, Chan KH, Nicholls J, et al. : Effective treatment of metastatic forms of Epstein-Barr virus-associated nasopharyngeal carcinoma with a novel adenovirus-based adoptive immunotherapy. Cancer Res 2012, 72:1116–1125. [DOI] [PubMed] [Google Scholar]
  • 38.Chia WK, Teo M, Wang WW, Lee B, Ang SF, Tai WM, Chee CL, Ng J, Kan R, Lim WT, et al. : Adoptive T-cell transfer and chemotherapy in the first-line treatment of metastatic and/or locally recurrent nasopharyngeal carcinoma. Mol Ther 2014, 22:132–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Huang J, Fogg M, Wirth LJ, Daley H, Ritz J, Posner MR, Wang FC, Lorch JH: Epstein-Barr virus-specific adoptive immunotherapy for recurrent, metastatic nasopharyngeal carcinoma. Cancer 2017, 123:2642–2650. [DOI] [PubMed] [Google Scholar]
  • 40.Saito M, Kono K: Landscape of EBV-positive gastric cancer. Gastric Cancer 2021, 24:983–989. [DOI] [PubMed] [Google Scholar]
  • 41.Johnson D, Ma BBY: Targeting the PD-1/ PD-L1 interaction in nasopharyngeal carcinoma. Oral Oncol 2021, 113:105127. [DOI] [PubMed] [Google Scholar]
  • 42.Goodman A, Patel SP, Kurzrock R: PD-1-PD-L1 immune-checkpoint blockade in B-cell lymphomas. Nat Rev Clin Oncol 2017, 14:203–220. [DOI] [PubMed] [Google Scholar]
  • 43.Minn AJ, Wherry EJ: Combination cancer therapies with immune checkpoint blockade: convergence on interferon signaling. Cell 2016, 165:272–275. [DOI] [PubMed] [Google Scholar]
  • 44.Shibata D, Weiss LM: Epstein-Barr virus-associated gastric adenocarcinoma. Am J Pathol 1992, 140:769–774. [PMC free article] [PubMed] [Google Scholar]
  • 45.Derks S, Liao X, Chiaravalli AM, Xu X, Camargo MC, Solcia E, Sessa F, Fleitas T, Freeman GJ, Rodig SJ, et al. : Abundant PD-L1 expression in Epstein-Barr virus-infected gastric cancers. Oncotarget 2016, 7:32925–32932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •46.Kawazoe A, Shitara K, Kuboki Y, Bando H, Kojima T, Yoshino T, Ohtsu, Ochiai A, Togashi Y, Nishikawa H, et al. : Clinicopathological features of 22C3 PD-L1 expression with mismatch repair, Epstein-Barr virus status, and cancer genome alterations in metastatic gastric cancer. Gastric Cancer 2019, 22:69–76. [DOI] [PubMed] [Google Scholar]; A clinically important study confirming that PD-L1 expression is more frequently found in EBV+ than EBV-metastatic gastric cancer, suggesting that EBV status is an important factor when assessing the potential clinical benefit of antiPD-1/PDL1 monoclonal antibody therapies.
  • 47.Panda A, Mehnert JM, Hirshfield KM, Riedlinger G, Damare S, Saunders T, Kane M, Sokol L, Stein MN, Poplin E, et al. : Immune activation and benefit from avelumab in EBV-positive gastric cancer. J Natl Cancer Inst 2018, 110:316–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hong M, Tang K, Qian J, Deng H, Zeng M, Zheng S, Ding K, Du Y, Sun R: Immunotherapy for EBV-associated nasopharyngeal carcinoma. Crit Rev Oncog 2018, 23:219–234. [DOI] [PubMed] [Google Scholar]
  • 49.Ooft ML, van Ipenburg JA, Braunius WW, Zuur CI, Koljenovic S, Willems SM: Prognostic role of tumor infiltrating lymphocytes in EBV positive and EBV negative nasopharyngeal carcinoma. Oral Oncol 2017, 71:16–25. [DOI] [PubMed] [Google Scholar]
  • 50.Zhao J, Guo C, Xiong F, Yu J, Ge J, Wang H, Liao Q, Zhou Y, Gong Q, Xiang B, et al. : Single cell RNA-seq reveals the landscape of tumor and infiltrating immune cells in nasopharyngeal carcinoma. Cancer Lett 2020, 477:131–143. [DOI] [PubMed] [Google Scholar]
  • 51.Leonetti A, Wever B, Mazzaschi G, Assaraf YG, Rolfo C, Quaini F, Tiseo M, Giovannetti E: Molecular basis and rationale for combining immune checkpoint inhibitors with chemotherapy in non-small cell lung cancer. Drug Resist Updat 2019, 46:100644. [DOI] [PubMed] [Google Scholar]
  • 52.Lee HM, Okuda KS, Gonzalez FE, Patel V: Current perspectives on nasopharyngeal carcinoma. Adv Exp Med Biol 2019, 1164:11–34. [DOI] [PubMed] [Google Scholar]
  • 53.Lv K, Li X, Yu H, Chen X, Zhang M, Wu X: Selection of new immunotherapy targets for NK/T cell lymphoma. Am J Transl Res 2020, 12:7034–7047. [PMC free article] [PubMed] [Google Scholar]
  • 54.Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, Schuster SJ, Millenson MM, Cattry D, Freeman GJ, et al. : PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med 2015, 372:311–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Armand P, Shipp MA, Ribrag V, Michot JM, Zinzani PL, Kuruvilla J, Snyder ES, Ricart AD, Balakumaran A, Rose S, et al. : Programmed death-1 blockade with pembrolizumab in patients with classical Hodgkin lymphoma after brentuximab vedotin failure. J Clin Oncol 2016, 34:3733–3739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Nakhoda S, Rizwan F, Vistarop A, Nejati R: Updates in the role of checkpoint inhibitor immunotherapy in classical Hodgkin’s lymphoma. Cancers 2022, 14:2936–2949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Takashima Y, Kawaguchi A, Sato R, Yoshida K, Hayano A, Homma J, Fukai J, Iwadate Y, Kajiwara K, Ishizawa S, et al. : Differential expression of individual transcript variants of PD-1 and PD-L2 genes on Th-1/Th-2 status is guaranteed for prognosis prediction in PCNSL. Sci Rep 2019, 9:10004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Qiu Y, Li Z, Pouzoulet F, Vishnu P, Copland JA 3rd, Knutson KL, Soussain C, Tun HW: Immune checkpoint inhibition by anti-PDCD1 (anti-PD1) monoclonal antibody has significant therapeutic activity against central nervous system lymphoma in an immunocompetent preclinical model. Br J Haematol 2018, 183:674–678. [DOI] [PubMed] [Google Scholar]
  • 59.Terziev D, Hutter B, Klink B, Stenzinger A, Stogbauer F, Glimm H, Frohling S, Wickenhauser C, Jordan K, Hurtz HJ, et al. : Nivolumab maintenance after salvage autologous stem cell transplantation results in long-term remission in multiple relapsed primary CNS lymphoma. Eur J Haematol 2018, 101:115–118. [DOI] [PubMed] [Google Scholar]
  • 60.Yiu SPT, Dorothea M, Hui KF, Chiang AKS: Lytic induction therapy against Epstein-Barr virus-associated malignancies: past, present, and future. Cancers 2020, 12:2142–2165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Andrei G, Trompet E, Snoeck R: Novel therapeutics for Epstein−Barr virus. Molecules 2019, 24:997–1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Stoker SD, Novalić Z, Wildeman MA, Huitema AD, Verkuijlen SA, Juwana H, Greijer AE, Tan IB, Middeldorp JM, de Boer JP: Epstein-Barr virus-targeted therapy in nasopharyngeal carcinoma. J Cancer Res Clin Oncol 2015, 141:1845–1857. [DOI] [PubMed] [Google Scholar]
  • ••63.Perrine SP, Hermine O, Small T, Suarez F, O’Reilly R, Boulad F, •• Fingeroth J, Askin M, Levy A, Mentzer SJ, et al. : A phase 1/2 trial of arginine butyrate and ganciclovir in patients with Epstein-Barr virus-associated lymphoid malignancies. Blood 2007, 109:2571–2578. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper shows the clinical potential of the ‘kick and kill’ combination approach of inducing lytic activation and inhibiting lytic replication.
  • 64.Faller DV, Mentzer SJ, Perrine SP: Induction of the Epstein-Barr virus thymidine kinase gene with concomitant nucleoside antivirals as a therapeutic strategy for Epstein-Barr virus-associated malignancies. Curr Opin Oncol 2001, 13:360–367. [DOI] [PubMed] [Google Scholar]
  • 65.Murata T, Tsurumi T: Switching of EBV cycles between latent and lytic states. Rev Med Virol 2014, 24:142–153. [DOI] [PubMed] [Google Scholar]
  • 66.Hui KF, Cheung AK, Choi CK, Yeung PL, Middeldorp JM, Lung ML, Tsao SW, Chiang AK: Inhibition of class I histone deacetylases by romidepsin potently induces Epstein-Barr virus lytic cycle and mediates enhanced cell death with ganciclovir. Int J Cancer 2016, 138:125–136. [DOI] [PubMed] [Google Scholar]
  • 67.Hui KF, Chiang AK: Suberoylanilide hydroxamic acid induces viral lytic cycle in Epstein-Barr virus-positive epithelial malignancies and mediates enhanced cell death. Int J Cancer 2010, 126:2479–2489. [DOI] [PubMed] [Google Scholar]
  • 68.Binne UK, Amon W, Farrell PJ: Promoter sequences required for reactivation of Epstein-Barr virus from latency. J Virol 2002, 76:10282–10289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Meng Q, Hagemeier SR, Fingeroth JD, Gershburg E, Pagano JS, Kenney SC: The Epstein-Barr virus (EBV)-encoded protein kinase, EBV-PK, but not the thymidine kinase (EBV-TK), is required for ganciclovir and acyclovir inhibition of lytic viral production. J Virol 2010, 84:4534–4542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Topalis D, Gillemot S, Snoeck R, Andrei G: Thymidine kinase and protein kinase in drug-resistant herpesviruses: heads of a lernaean hydra. Drug Resist Updat 2018, 37:1–16. [DOI] [PubMed] [Google Scholar]
  • 71.De Clercq E: Potential of acyclic nucleoside phosphonates in the treatment of DNA virus and retrovirus infections. Expert Rev Anti Infect Ther 2003, 1:21–43. [DOI] [PubMed] [Google Scholar]
  • •72.Drosu NC, Edelman ER, Housman DE: Tenofovir prodrugs potently inhibit Epstein-Barr virus lytic DNA replication by targeting the viral DNA polymerase. Proc Natl Acad Sci USA 2020, 117:12368–12374. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates that an approved nucleoside analogue inhibitor of HIV reverse transcriptase, Tenofovir, has potent and selective inhibitory activity for EBV DNA polymerase and prevents EBV lytic replication.
  • 73.Torkildsen O, Myhr KM, Skogen V, Steffensen LH, Bjornevik K: Tenofovir as a treatment option for multiple sclerosis. Mult Scler Relat Disord 2020, 46:102569. [DOI] [PubMed] [Google Scholar]
  • 74.Xie J, Wang Z, Fan W, Liu Y, Liu F, Wan X, Liu M, Wang X, Zeng D, Wang Y, et al. : Targeting cancer cell plasticity by HDAC inhibition to reverse EBV-induced dedifferentiation in nasopharyngeal carcinoma. Signal Transduct Target Ther 2021, 6:333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Yiu SPT, Hui KF, Munz C, Lo KW, Tsao SW, Kao RYT, Yang D, Chiang AKS: Autophagy-dependent reactivation of Epstein-Barr virus lytic cycle and combinatorial effects of autophagy-dependent and independent lytic inducers in nasopharyngeal carcinoma. Cancers 2019, 11:1871–1893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •76.Yiu SPT, Hui KF, Choi CK, Kao RYT, Ma CW, Yang D, • Chiang AKS: Intracellular iron chelation by a novel compound, C7, reactivates Epstein-Barr Virus (EBV) lytic cycle via the ERK-autophagy axis in EBV-positive epithelial cancers. Cancers 2018, 10:505–522. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper identifies a highly potent class of compounds that efficiently reactivate EBV in epithelial cell cancers through a mechanism involving iron chelation.
  • 77.Tikhmyanova N, Paparoidamis N, Romero-Masters J, Feng X, Mohammed FS, Reddy PAN, Kenney SC, Lieberman PM, Salvino JM: Development of a novel inducer for EBV lytic therapy. Bioorg Med Chem Lett 2019, 29:2259–2264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Tikhmyanova N, Tutton S, Martin KA, Lu F, Kossenkov AV, Paparoidamis N, Kenney S, Salvino JM, Lieberman PM: Small molecule perturbation of the CAND1-Cullin1-ubiquitin cycle stabilizes p53 and triggers Epstein-Barr virus reactivation. PLoS Pathog 2017, 13:e1006517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Tikhmyanova N, Schultz DC, Lee T, Salvino JM, Lieberman PM: Identification of a new class of small molecules that efficiently reactivate latent Epstein-Barr Virus. ACS Chem Biol 2014, 9:785–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Countryman JK, Gradoville L, Miller G: Histone hyperacetylation occurs on promoters of lytic cycle regulatory genes in Epstein-Barr virus-infected cell lines which are refractory to disruption of latency by histone deacetylase inhibitors. J Virol 2008, 82:4706–4719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Gradoville L, Kwa D, El-Guindy A, Miller G: Protein kinase C-independent activation of the Epstein-Barr virus lytic cycle. J Virol 2002, 76:5612–5626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Shah RR: Safety and tolerability of histone deacetylase (HDAC) inhibitors in oncology. Drug Saf 2019, 42:235–245. [DOI] [PubMed] [Google Scholar]
  • 83.Murata T, Sato Y, Kimura H: Modes of infection and oncogenesis by the Epstein-Barr virus. Rev Med Virol 2014, 24:242–253. [DOI] [PubMed] [Google Scholar]
  • 84.Bochkarev A, Barwell JA, Pfuetzner RA, Bochkareva E, Frappier L, Edwards AM: Crystal structure of the DNA-binding domain of the Epstein-Barr virus origin-binding protein, EBNA1, bound to DNA. Cell 1996, 84:791–800. [DOI] [PubMed] [Google Scholar]
  • 85.Bochkarev A, Barwell JA, Pfuetzner RA, Furey W Jr., Edwards AM, Frappier L: Crystal structure of the DNA-binding domain of the Epstein-Barr virus origin-binding protein EBNA 1. Cell 1995, 83:39–46. [DOI] [PubMed] [Google Scholar]
  • 86.Bochkarev A, Bochkareva E, Frappier L, Edwards AM: The 2.2 A structure of a permanganate-sensitive DNA site bound by the Epstein-Barr virus origin binding protein, EBNA1. J Mol Biol 1998, 284:1273–1278. [DOI] [PubMed] [Google Scholar]
  • 87.Jiang L, Lui YL, Li H, Chan CF, Lan R, Chan WL, Lau TC, Tsao GS, Mak NK, Wong KL: EBNA1-specific luminescent small molecules for the imaging and inhibition of latent EBV-infected tumor cells. Chem Commun 2014, 50:6517–6519. [DOI] [PubMed] [Google Scholar]
  • 88.Kim SY, Song KA, Kieff E, Kang MS: Small molecule and peptide-mediated inhibition of Epstein-Barr virus nuclear antigen 1 dimerization. Biochem Biophys Res Commun 2012, 424:251–256. [DOI] [PubMed] [Google Scholar]
  • •89.Jiang L, Xie C, Lung HL, Lo KW, Law GL, Mak NK, Wong KL: • EBNA1-targeted inhibitors: novel approaches for the treatment of Epstein-Barr virus-associated cancers. Theranostics 2018, 8:5307–5319. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper describes EBNA1 inhibitors based on the peptide sequence at the dimer-dimer interface. This unique targeting was shown to block EBV positive cancer cell growth in cell culture.
  • 90.Gianti E, Messick TE, Lieberman PM, Zauhar RJ: Computational analysis of EBNA1 “druggability” suggests novel insights for Epstein-Barr virus inhibitor design. J Comput Aided Mol Des 2016, 30:285–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Messick TE, Tolvinski L, Zartler ER, Moberg A, Frostell Å, Smith GR, Reitz AB, Lieberman PM: Biophysical screens identify fragments that bind to the viral DNA-binding proteins EBNA1 and LANA. Molecules 2020, 25:1760–1774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • ••92.Messick TE, Smith GR, Soldan SS, McDonnell ME, Deakyne JS, •• Malecka KA, Tolvinski L, van den Heuvel APJ, Gu BW, Cassel JA, et al. : Structure-based design of small-molecule inhibitors of EBNA1 DNA binding blocks Epstein-Barr virus latent infection and tumor growth. Sci Transl Med 2019, 11:eaau5612. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper describes the discovery, activity and selectivity of small molecule inhibitors of EBNA1 using a fragment-based approach. A Subsequent paper (93) demonstrated similar efficacy in EBVaGC. This class of small molecule has progressed to a clinical trial for treatment of NPC.
  • 93.Soldan SS, Anderson EM, Frase DM, Zhang Y, Caruso LB, Wang Y, Deakyne JS, Gewurz BE, Tempera I, Lieberman PM, et al. : EBNA1 inhibitors have potent and selective antitumor activity in xenograft models of Epstein-Barr virus-associated gastric cancer. Gastric Cancer 2021, 24:1076–1088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Griffin BD, Gram AM, Mulder A, Van Leeuwen D, Claas FH, Wang F, Ressing ME, Wiertz E: EBV BILF1 evolved to downregulate cell surface display of a wide range of HLA class I molecules through their cytoplasmic tail. J Immunol 2013, 190:1672–1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Fares S, Spiess K, Olesen ETB, Zuo J, Jackson S, Kledal TN, Wills MR, Rosenkilde MM: Distinct roles of extracellular domains in the Epstein-Barr virus-encoded BILF1 receptor for signaling and major histocompatibility complex class I downregulation. mBio 2019, 10:01707–01718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Knerr JM, Kledal TN, Rosenkilde MM: Molecular properties and therapeutic targeting of the EBV-encoded receptor BILF1. Cancers 2021, 13:4079–4095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •97.Kamara S, Guo Y,Mao S, Ye X, Li Q, Zheng M, Zhu J, Zhang J, Du W, Chen J, et al. : Novel EBV LMP1C-terminal domain binding affibody molecules as potential agents for in vivo molecular imaging diagnosis of nasopharyngeal carcinoma. Appl Microbiol Biotechnol 2021, 105:7283–7293. [DOI] [PubMed] [Google Scholar]; This study, along with ref 98, show the potential utility of affibodies derived from peptide display screening methods to bind LMP1 C-terminal domain and have utility in imaging of EBV+ NPC tumors.
  • 98.Zhu J, Kamara S, Cen D, Tang W, Gu M, Ci X, Chen J, Wang L, Zhu S, Jiang P, et al. : Generation of novel affibody molecules targeting the EBV LMP2A N-terminal domain with inhibiting effects on the proliferation of nasopharyngeal carcinoma cells. Cell Death Dis 2020, 11:213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Lo AK, Dawson CW, Lung HL, Wong KL, Young LS: The therapeutic potential of targeting BARF1 in EBV-associated malignancies. Cancers 2020, 12:1940–1952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Farrell CJ, Lee JM, Shin EC, Cebrat M, Cole PA, Hayward SD: Inhibition of Epstein-Barr virus-induced growth proliferation by a nuclear antigen EBNA2-TAT peptide. Proc Natl Acad Sci USA 2004, 101:4625–4630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Atkins SL, Motaib S, Wiser LC, Hopcraft SE, Hardy PB, Shackelford J, Foote P, Wade AH, Damania B, Pagano JS, et al. : Small molecule screening identifies inhibitors of the Epstein-Barr virus deubiquitinating enzyme, BPLF1. Antivir Res 2020, 173:104649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Cheng AZ, Yockteng-Melgar J, Jarvis MC, Malik-Soni N, Borozan I, Carpenter MA, McCann JL, Ebrahimi D, Shaban NM, Marcon E, et al. : Epstein-Barr virus BORF2 inhibits cellular APOBEC3B to preserve viral genome integrity. Nat Microbiol 2019, 4:78–88. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES