Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jun 1.
Published in final edited form as: Curr Treat Options Oncol. 2013 Jun;14(2):224–236. doi: 10.1007/s11864-013-0231-y

EBV Related Lymphomas: New Approaches to Treatment

Jennifer A Kanakry, Richard F Ambinder
PMCID: PMC3670765  NIHMSID: NIHMS463374  PMID: 23549980

Introduction

Over 90% of adults worldwide harbor lifelong latent EBV infection in a small fraction of their B-lymphocytes. While persistent latent EBV infection is common, the chance of any individual developing an EBV-related lymphoma is low and is modulated by ethnic, geographic, genetic, immunologic, and infectious cofactors. For example, in areas of holoendemic malaria, primary EBV infection is followed by a period of a few years where there is a propensity to develop EBV-positive Burkitt lymphoma (BL). The specific interplay of viral and malarial infection that leads to lymphoma remains poorly understood. In Europe, primary EBV infection that manifests as infectious mononucleosis is followed within 6 months to 10 years by increased risk of developing EBV-positive Hodgkin lymphoma (HL). Although both are B-lineage lymphomas, HL and BL are distinct, including differences in EBV latent gene expression. Whereas BL tumors typically express only a single viral protein Epstein-Barr nuclear antigen-1 (EBNA-1), HL tumors express EBNA-1 as well as viral latency membrane proteins (LMP)-1 and -2. These differences likely reflect differences in host factors and co-infection.[1]

EBV-related lymphomas are a heterogeneous group of hematologic malignancies but share the feature of harboring latent EBV within tumor cells. Certain lymphomas, such as endemic BL or HIV-associated primary central nervous system lymphoma, are EBV-positive in virtually 100% of cases. In HL, around 30% of cases in North America are EBV-positive. Diffuse large B-cell lymphoma (DLBCL) is rarely EBV-related, although cofactors such as chronic inflammation and perhaps immunosenescence of aging are associated with EBV-positivity. Immune dysfunction increases the likelihood of developing an EBV-related lymphoma, with high-risk groups including patients with HIV, congenital immunodeficiencies, post-transplant immunosuppression, or chronic active EBV. PTLD, particularly very early post-transplant, is usually EBV-positive.

There are different patterns of viral gene expression in different lymphomas, with associated ramifications for treatment. Only one or a few proteins may be expressed in a particular lymphoma type. The restricted protein expression allows the viral DNA to persist in cells, evading immune recognition. A corollary of this is that tumors with the least restricted viral gene expression patterns don’t occur in patients with functional immune systems. EBV-related lymphomas that arise in the setting of immunosuppression may respond favorably to therapies that ameliorate anti-viral immune function, such as reduction of immunosuppression or adoptive immunotherapy. By contrast, EBV-related tumors in immunocompetent individuals typically have more restricted viral gene expression and thus less immunogenicity, rendering the aforementioned therapies less effective. Some viral proteins activate proliferative pathways while others protect infected cells from apoptosis. Thus, it seems likely that viral gene expression modulates the effects of cytotoxic chemotherapy. Furthermore, antiviral therapies such as ganciclovir may be limited insofar as the agent requires phosphorylation for antiviral cytotoxic activity. The viral kinases required for ganciclovir phosphorylation are not typically expressed in tumors. Thus, these antiviral agents are not effective in the treatment of EBV-related lymphomas except perhaps in association with strategies to induce the viral kinases.

Nonetheless, the presence of virus in EBV-related lymphomas may be exploited for diagnostic and therapeutic purposes. Monitoring of EBV-DNA in peripheral blood is emerging as a marker of EBV-related lymphoma that may serve as a prognostic marker that could aid in risk-stratification, or guide response-adapted therapy decisions. Adoptive cellular immunotherapy targeting viral antigens is effective treatment in some settings. The development of strategies for altering viral gene expression patterns to facilitate targeting of virus-infected tumors continues.

TREATMENT

  • At present time, the EBV status of a lymphoma guides investigational therapies and the treatment of PTLD, particularly in the setting of allogeneic hematopoietic stem cell transplant (HSCT) where adoptive immunotherapy (either donor lymphocyte infusion or infusion of EBV-specific lymphocytes) has been shown to be effective.

Diet and lifestyle

  • There are no specific dietary or lifestyle risk factors for EBV-related lymphomas or lymphoproliferative disorders. However, there are areas of the world where certain EBV-related lymphomas have a much higher prevalence, such as EBV-positive BL in areas of Africa with holoendemic malaria, EBV-positive NK/T-cell lymphomas in parts of Asia, and EBV-positive HL in economically developing countries and among those of Hispanic ethnicity. The age of primary infection is a function of culture and diet but whether age of primary infection influences the overall incidence of EBV-related malignancies is not clear.

EBV-DNA Monitoring

  • There is growing interest in quantifying EBV-DNA in the peripheral blood as a surrogate marker for EBV-positive malignancies.[26]

  • While EBV-DNA is readily detected in the peripheral blood mononuclear cells (PBMCs) of even healthy, EBV-seropositive individuals, reflecting the small population of circulating latently infected B-lymphocytes, cell-free EBV-DNA (in plasma or serum) is not typically present in healthy individuals. Cell-free EBV-DNA can be derived from many sources, including dying EBV-infected tumor cells, dying latently infected benign cells, or virions. Particularly in immunocompromised hosts, cell-free EBV-DNA may be detectable for reasons apart from an underlying EBV-positive malignancy.

  • Some institutions monitor EBV-DNA levels in the blood of patients following allogeneic HSCT or solid organ transplantation (SOT).[7] However, the detection of EBV-DNA in blood specimens is not sufficient to diagnose EBV-PTLD.

  • In extranodal NK/T-cell lymphoma, EBV-DNA can be detected prior to therapy in the majority of patients. In one study, the treatment response rate was lower for patients with high levels of EBV-DNA in plasma prior to therapy; grade 4 treatment toxicities were also higher for these patients.[8] In extranodal NK/T-cell lymphoma, high pre-treatment plasma EBV-DNA levels correlated with higher International Prognostic Index scores and inferior survival outcomes.[3] Detectable post-treatment plasma EBV-DNA corresponded to inferior survival compared to those with undetectable post-treatment plasma EBV-DNA.[3]

  • In HL, there is a strong correlation between EBV-positive tumors and elevated cell-free EBV-DNA.[4, 5] One study showed that high EBVDNA in plasma of HL patients corresponds to higher stage disease and higher International Prognostic scores.[6] Several studies have demonstrated that cell-free EBV-DNA corresponds to active EBV-positive disease, where patients with EBV-positive HL typically have detectable EBV-DNA in their blood at diagnosis or in the setting of relapsed/refractory disease while patients with EBV-negative HL tumors or EBV-positive tumors in remission will not have EBV-DNA in their blood.[4] In a trial of rituximab-ABVD for patients with advanced Hodgkin lymphoma, those with EBV-positive tumors experienced rapid declines in EBV-DNA viral load after one cycle of therapy, which corresponded to treatment responses in all cases.[9]

Pharmacologic treatment

  • In most instances, the approach to EBV-positive lymphomas does not differ from EBV-negative lymphomas of the same histology. The exceptions are in the context of investigational protocols or where an adoptive immunotherapy approach is available.

  • When EBV-positive lymphomas arise in the setting of immunosuppression, ameliorating the immune defect can assist in the treatment of these lymphomas. In HIV-associated lymphomas, antiretroviral therapy is generally appropriate although potential drug interactions and the effects of chemotherapy on the ability to maintain HAART therapy in terms of nausea, vomiting and mucositis must be considered with regard to the timing of antiretroviral therapy. However, for EBV-related lymphomas in HIV patients, initiation of antiretroviral therapy alone is inadequate for treatment. This is in contrast to AIDS-associated Kaposi sarcoma where initiation of antiretroviral therapy is often a standard approach in patients who are asymptomatic or minimally symptomatic and are antiretroviral naïve. In EBV-PTLD, select cases may benefit from reduction of immunosuppression as the sole intervention or as part of the treatment plan.

Rituximab

For the treatment of EBV-PTLD, rituximab as a single agent has a reported efficacy of 63% on a systematic review and is considered first-line treatment by many transplant centers.[10] The benefits of rituximab for EBVPTLD are that many patients can be spared the toxicities of combination chemotherapy and that their immunosuppression can be often be continued, leading to lower risk of graft rejection and/or GVHD. Rituximab may be effective in EBV-PTLD not only because it targets the CD20(+) tumor, but also because, through B-cell depletion, it may shift the ratio of EBV-infected B-lymphocytes to EBV-specific cytotoxic T-cells in favor of an antiviral/ anti-tumor immune response. The benefit (or detriment) of harboring EBV within other lymphomas, as it relates to rituximab efficacy, has not been studied. There is some evidence that LMP-1, via activation of downstream signaling pathways, may alter the sensitivity of EBV-positive lymphomas to rituximab. One recent study showed that rituximab-sensitive EBV-positive lymphoma cells were rendered resistant to rituximab treatment after transfection with LMP-1.[11] Further, LMP-1 transfection induced Akt phosphorylation and Akt inhibition restored rituximab sensitivity of these cells.[11]

Standard dosage: Variable depending on regimen

Contraindications: None, precautions if history of infusion reaction

Main drug interactions: None, although concurrent use with live vaccines may increase risk of infection by the live vaccine.

Main side effects: Infusion reactions, generally mild, can occur. Hypogammaglobulinemia or lateonset neutropenia are recognized to occur in small numbers of patients.

Special points: Extranodal lymphomatous involvement, including the gastrointestinal tract, is often a feature of EBV-associated lymphomas and gastrointestinal perforation has been reported with rituximab.

Cost/cost-effectiveness: Average wholesale price of rituximab is $582.19 per 10 mL (100 mg) vials. At the average dose of 800 mg, the cost of one infusion would be $4,657.52.

Reduction or Modification of Immunosuppression

Reduction of pharmacologic immunosuppression (RI) can be an effective approach to the treatment of EBV-PTLD in some cases, although this strategy must be balanced against the risk of transplant rejection (or GVHD in HSCT). Furthermore, the rapidity with which an EBV-specific immune response will regenerate following RI should be considered. In settings where T-cells have been removed either by T-cell depletion of a stem cell graft as in HSCT or following treatment of the patient with antibodies that specifically target T-cells such as OKT3, antithymocyte globulin, or alemtuzumab, immune reconstitution may be delayed for weeks even after the immunosuppressive agents have been discontinued. In some settings, particularly SOT, where immunosuppression is a result of calcineurin inhibitors and antimetabolites, recovery of anti-viral immune function following RI may occur more rapidly.

As an alternative to RI, modification of the immunosuppression regimen to include agents that have potential anti-tumor and anti-viral properties is an attractive option, allowing treatment of the EBV-PTLD while maintaining the level of immunosuppression necessary to prevent graft rejection and GVHD. Immunosuppressive agents such as mammalian target of rapamycin (mTOR) inhibitors have been explored in PTLD given their antitumor properties in other settings. The mTOR signaling pathway is important in B-cell proliferation and early EBV-PTLD lesions have been shown to have activation of the mTOR pathway.[12, 13] Remissions of EBV-PTLD have been reported in SOT patients upon modification of their immunosuppressant regimen to include an mTOR inhibitor instead of a calcineurin inhibitor.[14, 15] In vitro, rapamycin has been shown to inhibit the proliferation of EBV-positive lymphoma cell lines, but not EBV-negative lymphoma cell lines, suggesting possible anti-viral activity as well.[16]

Surgery and Interventional Procedures

  • In the setting of PTLD involving a transplanted kidney or other localized disease, surgery to remove lymphoma is sometimes undertaken, particularly if RI has failed.

  • Excisional biopsy is valuable in the diagnosis of lymphoma and for performing appropriate virally-related tissue evaluations, including EBV determination by in situ hybridization for highly-expressed EBV RNAs (EBER) or immunohistochemistry for LMP-1.

Other treatments

  • Adoptive cellular immunotherapy

Adoptive Cellular Immunotherapy

Adoptive T-cell therapy with EBV-specific cytotoxic T-cells (EBV-CTLs) has been used for the treatment or prevention of EBV-PTLD since 1995 and has proven to be safe and effective, even in patients with refractory or relapsed disease.[17, 18] Clinical outcomes of patients with EBV-PTLD treated with EBV-CTLs were recently reviewed, with responses seen in the majority of patients.[19] Among 101 HSCT patients who were deemed highrisk for EBV-PTLD, none developed EBV-PTLD after prophylactic treatment with EBV-CTLs.[17] This same group reported that 11 of 13 patients treated for established EBV-PTLD achieved durable complete remissions. In one series, over two-thirds of HSCT patients with EBV-PTLD had durables remissions following infusion of EBV-CTLs with no cases of GVHD (compared to 17% in patients receiving HLA-matched donor lymphocyte infusion for EBV-PTLD).[ 20]

EBV-CTLs can be expanded ex vivo using lymphoblastoid cell lines (LCLs), a process that requires repetitive antigen stimulation and typically takes 4–12 weeks. However, EBV-PTLD is often aggressive, necessitating urgent treatment. A group in Edinburgh has made efforts to make EBV-CTLs an “off the shelf” product by creating a bank of cryopreserved EBV-CTLs expanded from healthy blood donors.[21] The group has created a bank of EBV-CTLs from donors with various HLA types, making products available for quick matching if a patient develops EBVPTLD. An overall response rate of 48% was observed with these banked allogeneic EBV-CTLs when given to SOT patients with EBV-PTLD. The survival of third-party EBV-CTLs is hindered by allogeneic responses and their efficacy may be limited by ongoing immunosuppressive therapy. Nonetheless, the approach has promise and follow-up investigations are ongoing. Others have reported that overnight stimulation is adequate to generate EBNA-1-specific CTLs from peripheral blood mononuclear cells using EBNA-1-specific interferon gamma secretion as a selection marker.[22] Adoptive therapy with the resultant cellular products led to in vivo expansion of EBVCTLs in 80% of patients and clinical responses in 70% of patients.[22] Similar approaches to the generation of cytomegalovirus (CMV)-specific and adenovirus-specific CTLs are ongoing and there has been exploration of multivirus- specific (EBV, CMV, adenovirus) CTLs in HSCT patients receiving T-cell depleted grafts.[23, 24]

Engineering specific CTLs has attracted considerable attention. In the HSCT setting, EBV-CTLs have been shown to persist in vivo long-term, demonstrated by detection of gene-marked cells many years after infusion.[17] In the SOT setting where patients are often maintained on immunosuppression for years or for life, EBV-CTLs have been engineered to be resistant to calcineurin inhibitors so as to enhance survival and efficacy.[25, 26] Conversely, concerns that CTLs might induce GVHD have led to the development of cellular products that are engineered with suicide genes, so that in the event that these cells lead to GVHD, the cells may be turned off pharmacologically.[27, 28]

Successes in EBV-PTLD spurred interest in the use of EBV-CTLs for patients with other EBV-related diseases, including EBV-positive HL and EBV-positive undifferentiated nasopharyngeal carcinoma (NPC).[29] One of the barriers to treatment outside the HSCT setting is achieving in vivo expansion of infused cells. A variety of approaches have been explored. Among them, short-term lymphodepletion with anti-CD45 monoclonal antibodies to facilitate in vivo expansion of EBV-specific CTLs in NPC patients has been followed by adoptive immunotherapy.[30] When the approach was applied, increases in EBV-CTLs were seen, with some patients achieving clinical remission. In vivo persistence of EBV-CTLs correlated with tumor regression and improved clinical outcomes.[30] In pre-clinical experiments, IL-15 has been shown to support in vivo proliferation of EBVCTLs that were initially expanded ex vivo, even in the presence of functional regulatory T-cells.[31] Other investigations involve engineering CTLs to be resistant to tumor-derived immunomodulatory cytokines such as TGF-β that are believed to be important components of the tumor milieu in HL.[32] The viral proteins LMP-1 and LMP-2 appear to drive the transcriptional activation of galectin-1 (Gal1) in HL tumors.[33] It has been suggested that Gal1 expression skews the immune response away from a cytotoxic T-cell response and towards a tolerogenic, regulatory T-cell response, potentially facilitating the tumor immune escape in HL.[34] EBV-CTLs are sensitive to Gal1-mediated apoptosis and Gal1 neutralizing antibodies have been shown to protect EBV-CTLs from Gal1 cytotoxicity.[33] Gal1 neutralizing antibodies may have a future role in the immunotherapeutic approaches to EBV-positive lymphomas.

EBV-specific T-cells are also being investigated as novel therapies for non-EBV associated hematologic malignancies, such as chronic lymphocyte leukemia. In an ongoing clinical trial, anti-CD19 antibodies are joined to T-cells as form a chimeric receptor. In attempts to increase longevity of the chimeric receptor-T-cell, half will have a CD28 protein attached and half will be selectively expanded to recognize EBV. In a similar design, there is a Phase I clinical trial for patients with CD30-posiitve lymphomas where an anti-CD30 antibody is joined to EBV-CTLs.

Standard dosage: Use is investigational

Contraindications: Per clinical trial criteria, but often allogeneic HSCT recipients with GVHD of grade 2 or greater are considered ineligible

Main drug interactions: Some immunosuppressive therapies may hinder CTL expansion in vivo

Main side effects: Localized swelling at sites of EBV-PTLD with inflammation and necrosis. Some patients have had GVHD or recurrence of GVHD.

Special points: Limitations include production time, availability of donors outside of HSCT setting, concerns about GVHD, manufacturing cells limited to certain centers

Cost/cost-effectiveness: In 2009, the reported cost of manufacturing and infusing EBV-CTLs at an established facility was reported to be $6,095[17]

Emerging therapies

  • Signaling pathway inhibitors

  • Lytic inducers coupled with antiherpesvirus agents

  • EBV vaccines

Signaling Pathway Inhibitors

The EBV protein LMP-2, which is present in some EBV-related lymphomas, mimics a functional B-cell receptor (BCR) and may activate downstream signaling pathways, including Syk, Akt, and mTOR. The Lyn/Syk pathway is downstream of the BCR and, when activated, functions to promote the survival of B-lymphocytes. In murine NHL tumors, Syk is required for tumor survival in vitro and, in vivo, tumors that do not express Syk have increased apoptosis.[35] Syk signaling leads to activation of Akt and XIAP, a caspase inhibitor, resulting in inhibition of apoptosis.[36] Inhibition of Syk leads to tumor regression in NHL cell lines and mouse models.[35, 37, 38] Syk inhibitors have clinical anti-tumor activity that has been demonstrated in patients with CLL and NHL.[3941] In LMP-2A transgenic mice, B-cells can survive and proliferate despite lacking functional BCRs.[42] In LMP-2A/myc double transgenic mice, lymphoid tumors develop, suggesting that LMP-2A functions to allow cells with aberrant myc expression to escape apoptotic signals and survive.[43] Among the Src family kinases, Lyn kinase in particular associates with LMP-2A, leading to activation of B-cell survival signaling pathways that bypass signaling through the BCR.[44] Dasatinib has been shown to inhibit the proliferation of LMP-2A-expressing B-lymphocytes in vitro, as well as splenomegaly and lymphomagenesis in LMP-2A/myc double transgenic mice.[45] The mechanism of action of dasatinib appears to be related to the inhibition of LMP-2A-induced Lyn signaling.[45] In EBV-PTLD cell lines, inhibition of Syk led to reduced tumor proliferation and induced apoptosis.[36] As more small molecule inhibitors become available, evaluating these agents in EBV-related lymphomas may prove effective beyond the anti-tumor activity seen in EBV-negative lymphomas, due to the viral association.

The EBV protein LMP-1, a mimic of CD40, is recognized to activate the PI3K/Akt pathway through noncanonical Syk signaling.[4648] LMP-1 has been shown to increase the localization of PI3K to lipid rafts, with cholesterol depletion resulting in inhibition of PI3K localization and decreased Akt activation.[49] In extranodal NK/T-cell lymphoma, tumor LMP-1 expression, determined by immunohistochemistry on tumor tissue, correlated with Akt phosphorylation, more localized disease, and better overall survival.[50] In EBV-positive NPC, the PI3K/Akt and mTOR pathways are often activated.[5153] PI3K inhibitors, Akt inhibitors, and dual PI3K/mTOR inhibitors are all in development. Akt inhibitors, such as MK-2206, have been shown to inhibit growth of NPC cell lines.[54] MK-2206 is currently being investigated in Phase II clinical trials for patients with relapsed/refractory HL and NHL, as well as recurrent NPC. LMP-1, by signaling through the JAK/STAT pathway, may also function to increase programmed cell death ligand 1 (PD-L1) expression in EBV-positive tumors, including EBV-PTLD and EBV-positive HL.[55] EBV-transformed LCLs were shown to have high levels of JAK3-associated STAT proteins (pSTAT3 and pSTAT5) and enhanced LMP-1 increased STAT5 activity. JAK3 inhibition led to decreased PD-L1 expression in EBV-transformed LCLs. PD-L1 is a cosignaling molecule that inhibits the function of activated effector T-cells. PD-L1 thus acts to dampen T-cell mediated immune responses, including anti-tumor immunity. PD-1 blockade is a promising targeted therapy in hematologic malignancies and has already proven to be effective in the treatment of other solid tumor malignancies. PD-L1 blockade and reactivation of a dampened immune response may be especially relevant for EBV-related tumors.

Lytic Inducers Coupled With Antiherpesvirus Agents

To persist for the lifetime of the host, EBV maintains tight control over the switch between latent and lytic gene expression, with latency characterized by very restricted gene expression. Lytic proteins, such as viral thymidine kinase, are not expressed during latency. As a result, antiherpesvirus drugs that rely on phosphorylation by viral thymidine kinase for conversion of the prodrug to its active form, are not effective during latent infection. Thus, there is interest in the pharmacologic lytic activation of EBV to render infected tumor cells susceptible to antiviral agents such as ganciclovir. Furthermore, lytic activation in itself may render the infected cells more susceptible to immune recognition and killing due to a less restricted expression of viral proteins.

Numerous therapies induce lytic viral activation to varying degrees, including chemotherapeutic agents, radiation, steroids, histone deacetylase inhibitors (HDACi), and others. Lytic activation may be part of the DNA damage response or unfolded protein response to endoplasmic reticulum stress. Bortezomib, a proteasome inhibitor, has been shown to activate EBV lytic gene expression in EBV-positive BL cell lines.[56] EBV-positive NPC cell lines treated with arsenic trioxide showed increased lytic gene expression and decreased viability.[57] When ganciclovir was added to arsenic-treated EBV-positive cells, viability was further reduced while EBV-negative cells showed no changes in viability after treatment with arsenic alone, ganciclovir alone, or the combination.[57] LMP-1-positive cells were also shown to have higher levels of promyelocytic leukemia nuclear bodies (PML-NB) protein expression and treatment with arsenic decreased PML-NB protein levels.[57] PML-NB degradation may have an important role in the switch from latent to lytic herpesvirus gene expression and LMP-1 may increase PML-NB expression to maintain latency.[57] EBNA-1 may also mediate the switch between latency and lytic activation through alterations in PML-NB.[58] Viral genes expressed at early stages of the lytic cycle have also been found to participate in PML-NB disruption.[59]

HDACis have been shown to induce lytic gene expression and sensitize EBV-positive lymphoma cell lines to ganciclovir at drug concentrations achievable in patients.[6062] In EBV-positive NPC cell lines, gemcitabine and valproic acid had a synergistic effect on lytic activation.[62] The addition of ganciclovir increased cytotoxic activity compared to treatment with the lytic inducers alone, which was not observed in EBV-negative cell lines.[62] Notably, the production of viral capsid proteins and new virions were also inhibited.[62] In a patient with a refractory EBV-positive DLBCL, treatment with valproic acid and ganciclovir was associated with a rise in EBVDNA in the plasma that was found to be unencapsidated by DNAse assay, suggesting that the rise in EBV-DNA was tumor-derived.[63] In a clinical trial of 15 patients with refractory EBV-positive lymphomas, 10 responded to treatment with arginine butyrate, an HDACi and lytic inducer, in combination with ganciclovir.[64] However, it remains unclear if the response seen was due to the anti-tumor activity of the HDACi or truly related to lytic induction and ganciclovir sensitization of tumor cells. Epigenetic modifying agents such as 5-azacytidine also induce lytic gene expression in EBV-positive cell lines and, when combined with ganciclovir, lead to caspasemediated apoptosis.[65] Parthenolide, a plant extract, has been shown to induce lytic gene expression in EBV-positive BL cell lines and to have cytotoxic activity that synergizes with ganciclovir.[66]

The ataxia-telangiectasia-mutated (ATM) kinase plays a role in EBV lytic induction, with ATM kinase activity required for agents such as bortezomib, HDACis, and others to effectively induce lytic gene expression.[67] Treatment of EBV-positive cell lines with an ATM inhibitor decreased lytic gene expression, with similar findings in an ATM knockdown model.[67] Drugs which activate ATM appear to result in lytic induction.[67]

EBV Vaccines

A recombinant glycoprotein gp350 vaccine in healthy volunteers appears to reduce the incidence of symptomatic infectious mononucleosis but may not prevent primary infection.[68, 69] Whether such vaccination would impact on the incidence of EBV-related malignancies is unknown. A peptide epitope-based vaccine aimed at stimulating T-cell mediated immunity against immunodominant latency antigens has been shown to induce EBV-specific T-cell responses in healthy volunteers and may have a role in the prevention of primary infection, or in the prevention or treatment of EBV-related malignancies.[70, 71] In a trial of pediatric patients awaiting SOT, the gp350 vaccine was given pre-transplant with the hopes of improving anti-viral T-cell immunity to prevent EBVPTLD.[ 72] However, the gp350 vaccine failed to elicit a durable immune response in the majority of trial participants.[72] Phase I EBV vaccine trials have been completed in patients with EBV-positive malignancies in remission using a modified vaccinia Ankara EBNA-1/LMP-1 vaccine.

Pediatric considerations

  • Pediatric patients are more likely to be EBV-seronegative, which is a consideration in pediatric transplant patients, where the risk of EBVPTLD is heightened in these circumstances.

Opinion statement.

In the treatment of Epstein-Barr virus (EBV)-related lymphomas, there are few therapies specifically targeted against the latent virus within these tumors; in most cases the treatment approach is not different than the approach to EBV-negative lymphomas. Nonetheless, current and emerging therapies focused on exploiting aspects of EBV biology may offer more targeted strategies for EBV-positive lymphomas in the future. Conceptually, EBV-specific approaches include bolstering the anti-viral/ anti-tumor immune response with vaccines or EBV-specific cytotoxic T-lymphocytes, activating lytic viral genes to render the tumor cells susceptible to antiviral therapies, and inhibiting the downstream pro-survival or anti-apoptotic pathways that may be activated by latent EBV proteins.

EBV-specific cytotoxic T-cell infusions have proven effective in EBV-related posttransplantation lymphoproliferative disorder (EBV-PTLD) and expanding such adoptive immunotherapies to other EBV-related malignancies is an area of active research. However, other EBV-related lymphomas typically have more restricted, less immunogenic arrays of viral antigens to therapeutically target with adoptive immunotherapy compared to EBV-PTLD. Furthermore, the malignant EBV-positive tumor cells of Hodgkin lymphoma are scattered amid a dense infiltrate of regulatory T-cells, macrophages, and other cells that may dampen the anti-tumor efficacy of adoptive immunotherapy. Strategies to overcome these obstacles are areas of ongoing pre-clinical and clinical investigations.

Some emerging approaches to EBV-related lymphomas include the coupling of agents that induce lytic viral replication with anti-herpesvirus agents, or the use of small molecule inhibitors that block signaling pathways that are constitutively activated by EBV. EBV vaccines seem most promising in the treatment or prevention of EBV-related malignancies, rather than the prevention of primary EBV infection. EBV vaccine trials in patients with residual or low-bulk EBV-related malignancies or for the prevention of EBV-PTLD in EBV-seronegative patients awaiting solid organ transplantation are ongoing.

Footnotes

Conflicts of Interest:

Jennifer A. Kanakry declares that she has no conflicts of interest.

Richard F. Ambinder declares that he has no conflicts of interest.

References and Recommended Reading

Papers of particular interest, published recently, have been highlighted as:

• Of importance

•• Of major importance

  • 1.Levin LI, Chang ET, Ambinder RF, et al. Atypical prediagnosis Epstein-Barr virus serology restricted to EBV-positive Hodgkin lymphoma. Blood. 2012;120:3750–3755. doi: 10.1182/blood-2011-12-390823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Jones K, Nourse JP, Keane C, et al. Tumor-specific but not nonspecific cell-free circulating DNA can be used to monitor disease response in lymphoma. Am J Hematol. 2012;87:258–265. doi: 10.1002/ajh.22252. [DOI] [PubMed] [Google Scholar]
  • 3. Wang ZY, Liu QF, Wang H, et al. Clinical implications of plasma Epstein-Barr virus DNA in early-stage extranodal nasal-type NK/T-cell lymphoma patients receiving primary radiotherapy. Blood. 2012;120:2003–2010. doi: 10.1182/blood-2012-06-435024. Elevated pre-treatment EBV-DNA copy number in plasma is associated with inferior survival outcomes in extranodal NK/T-cell lymphoma.
  • 4.Gandhi MK, Lambley E, Burrows J, et al. Plasma Epstein-Barr virus (EBV) DNA is a biomarker for EBV-positive Hodgkin's lymphoma. Clin Cancer Res. 2006;12:460–464. doi: 10.1158/1078-0432.CCR-05-2008. [DOI] [PubMed] [Google Scholar]
  • 5. Kanakry JA, Li H, Gellert LL, et al. Plasma Epstein-Barr virus DNA as a marker of tumor response in Hodgkin lymphoma. 2012 Abstract #8003. Plasma EBV-DNA correlates well with EBV tumor status and pre-treatment plasma EBV-DNA positivity is an independent prognostic marker of inferior progression-free survival in Hodgkin lymphoma.
  • 6. Hohaus S, Santangelo R, Giachelia M, et al. The viral load of Epstein-Barr virus (EBV) DNA in peripheral blood predicts for biological and clinical characteristics in Hodgkin lymphoma. Clin Cancer Res. 2011;17:2885–2892. doi: 10.1158/1078-0432.CCR-10-3327. High plasma EBV-DNA correlates with tumor-infiltrating macrophages in Hodgkin lymphoma.
  • 7.Styczynski J, Reusser P, Einsele H, et al. Management of HSV, VZV and EBV infections in patients with hematological malignancies and after SCT: guidelines from the Second European Conference on Infections in Leukemia. Bone Marrow Transplant. 2009;43:757–770. doi: 10.1038/bmt.2008.386. [DOI] [PubMed] [Google Scholar]
  • 8. Ito Y, Kimura H, Maeda Y, et al. Pretreatment EBV-DNA copy number is predictive of response and toxicities to SMILE chemotherapy for extranodal NK/T-cell lymphoma, nasal type. Clin Cancer Res. 2012;18:4183–4190. doi: 10.1158/1078-0432.CCR-12-1064. Plasma EBV-DNA copy number predicts response to chemotherapy and grade 4 toxicity for patients with extranodal NK/T-cell lymphoma.
  • 9.Kasamon YL, Jacene HA, Gocke CD, et al. Phase 2 study of rituximab-ABVD in classical Hodgkin lymphoma. Blood. 2012;119:4129–4132. doi: 10.1182/blood-2012-01-402792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Styczynski J, Einsele H, Gil L, Ljungman P. Outcome of treatment of Epstein-Barr virus-related post-transplant lymphoproliferative disorder in hematopoietic stem cell recipients: a comprehensive review of reported cases. Transpl Infect Dis. 2009;11:383–392. doi: 10.1111/j.1399-3062.2009.00411.x. [DOI] [PubMed] [Google Scholar]
  • 11. Kim JH, Kim WS, Park C. Epstein-Barr virus latent membrane protein-1 protects B-cell lymphoma from rituximab-induced apoptosis through miR-155-mediated Akt activation and up-regulation of Mcl-1. Leuk Lymphoma. 2012;53:1586–1591. doi: 10.3109/10428194.2012.659736. Role of Akt pathway in LMP-1-mediated rituximab resistance in EBV-positive lymphomas.
  • 12. Nelson BP, Wolniak KL, Evens A, Chenn A, Maddalozzo J, Proytcheva M. Early posttransplant lymphoproliferative disease: clinicopathologic features and correlation with mTOR signaling pathway activation. Am J Clin Pathol. 2012;138:568–578. doi: 10.1309/AJCPQYYE04AVGVYI. mTOR activation in EBV-PTLD.
  • 13.El-Salem M, Raghunath PN, Marzec M, et al. Constitutive activation of mTOR signaling pathway in post-transplant lymphoproliferative disorders. Lab Invest. 2007;87:29–39. doi: 10.1038/labinvest.3700494. [DOI] [PubMed] [Google Scholar]
  • 14.Gibelli NE, Tannuri U, Pinho-Apezzato ML, et al. Sirolimus in pediatric liver transplantation: a single-center experience. Transplant Proc. 2009;41:901–903. doi: 10.1016/j.transproceed.2009.01.054. [DOI] [PubMed] [Google Scholar]
  • 15.Garcia VD, Bonamigo Filho JL, Neumann J, et al. Rituximab in association with rapamycin for post-transplant lymphoproliferative disease treatment. Transpl Int. 2003;16:202–206. doi: 10.1007/s00147-002-0487-9. [DOI] [PubMed] [Google Scholar]
  • 16.Vaysberg M, Balatoni CE, Nepomuceno RR, Krams SM, Martinez OM. Rapamycin inhibits proliferation of Epstein-Barr virus-positive B-cell lymphomas through modulation of cell-cycle protein expression. Transplantation. 2007;83:1114–1121. doi: 10.1097/01.tp.0000260142.38619.9c. [DOI] [PubMed] [Google Scholar]
  • 17. Heslop HE, Slobod KS, Pule MA, 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: 10.1182/blood-2009-08-239186. Long-term outcomes of EBV-specific CTL therapy for the prevention and treatment of EBV-PTLD.
  • 18.Rooney CM, Smith CA, Ng CYC, et al. Use of Gene-Modified Virus-Specific T-Lymphocytes to Control Epstein-Barr-Virus-Related Lymphoproliferation. Lancet. 1995;345:9–13. doi: 10.1016/s0140-6736(95)91150-2. [DOI] [PubMed] [Google Scholar]
  • 19. Bollard CM, Rooney CM, Heslop HE. T-cell therapy in the treatment of post-transplant lymphoproliferative disease. Nat Rev Clin Oncol. 2012;9:510–519. doi: 10.1038/nrclinonc.2012.111. Review of EBV-CTL therapies.
  • 20. Doubrovina E, Oflaz-Sozmen B, Prockop SE, 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: 10.1182/blood-2011-08-371971. Comparison of clinical outcomes following donor lymphocyte infusion versus EBV-CTL therapy for post-HSCT EBV-PTLD.
  • 21.Haque T, Wilkie GM, Jones MM, et al. Allogeneic cytotoxic T-cell therapy for EBV-positive posttransplantation lymphoproliferative disease: results of a phase 2 multicenter clinical trial. Blood. 2007;110:1123–1131. doi: 10.1182/blood-2006-12-063008. [DOI] [PubMed] [Google Scholar]
  • 22. Icheva V, Kayser S, Wolff D, 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. 2012 doi: 10.1200/JCO.2011.39.8495. Interferon-gamma capture technique for rapid isolation of EBV-CTLs.
  • 23.Feuchtinger T, Opherk K, Bethge WA, et al. Adoptive transfer of pp65-specific T cells for the treatment of chemorefractory cytomegalovirus disease or reactivation after haploidentical and matched unrelated stem cell transplantation. Blood. 2010;116:4360–4367. doi: 10.1182/blood-2010-01-262089. [DOI] [PubMed] [Google Scholar]
  • 24. Gerdemann U, Keirnan JM, Katari UL, et al. Rapidly generated multivirus-specific cytotoxic T lymphocytes for the prophylaxis and treatment of viral infections. Mol Ther. 2012;20:1622–1632. doi: 10.1038/mt.2012.130. Use of synthetic peptides and cytokines to facilitate the rapid expansion of multi-virus-specific CTLs.
  • 25.De Angelis B, Dotti G, Quintarelli C, et al. Generation of Epstein-Barr virus-specific cytotoxic T lymphocytes resistant to the immunosuppressive drug tacrolimus (FK506) Blood. 2009;114:4784–4791. doi: 10.1182/blood-2009-07-230482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Brewin J, Mancao C, Straathof K, et al. Generation of EBV-specific cytotoxic T cells that are resistant to calcineurin inhibitors for the treatment of posttransplantation lymphoproliferative disease. Blood. 2009;114:4792–4803. doi: 10.1182/blood-2009-07-228387. [DOI] [PubMed] [Google Scholar]
  • 27.Quintarelli C, Savoldo B, Dotti G. Gene therapy to improve function of T cells for adoptive immunotherapy. Methods Mol Biol. 2010;651:119–130. doi: 10.1007/978-1-60761-786-0_8. [DOI] [PubMed] [Google Scholar]
  • 28.Marin V, Cribioli E, Philip B, et al. Comparison of different suicide gene strategies for the safety improvement of genetically manipulated T cells. Hum Gene Ther Methods. 2012 doi: 10.1089/hgtb.2012.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.He J, Tang XF, Chen QY, et al. Ex vivo expansion of tumor-infiltrating lymphocytes from nasopharyngeal carcinoma patients for adoptive immunotherapy. Chin J Cancer. 2012;31:287–294. doi: 10.5732/cjc.011.10376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Louis CU, Straathof K, Bollard CM, et al. Enhancing the in vivo expansion of adoptively transferred EBV-specific CTL with lymphodepleting CD45 monoclonal antibodies in NPC patients. Blood. 2009;113:2442–2450. doi: 10.1182/blood-2008-05-157222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Perna SK, De Angelis B, Pagliara D, et al. Interleukin 15 provides relief to cytotoxic T lymphocytes from regulatory T cell-mediated inhibition: implications for adoptive T-cell based therapies of lymphoma. Clin Cancer Res. 2012 doi: 10.1158/1078-0432.CCR-12-2143. Role of IL-15 in promoting the survival and expansion of EBV-CTLs in the presence of regulatory T-cells.
  • 32.Foster AE, Dotti G, Lu A, et al. Antitumor activity of EBV-specific T lymphocytes transduced with a dominant negative TGF-beta receptor. J Immunother. 2008;31:500–505. doi: 10.1097/CJI.0b013e318177092b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Ouyang J, Juszczynski P, Rodig SJ, et al. Viral induction and targeted inhibition of galectin-1 in EBV+ posttransplant lymphoproliferative disorders. Blood. 2011;117:4315–4322. doi: 10.1182/blood-2010-11-320481. Role of galectin-1 in EBV-positive lymphomas and the EBV-specific CTL immune response.
  • 34.Juszczynski P, Ouyang J, Monti S, et al. The AP1-dependent secretion of galectin-1 by Reed Sternberg cells fosters immune privilege in classical Hodgkin lymphoma. Proc Natl Acad Sci U S A. 2007;104:13134–13139. doi: 10.1073/pnas.0706017104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Young RM, Hardy IR, Clarke RL, et al. Mouse models of non-Hodgkin lymphoma reveal Syk as an important therapeutic target. Blood. 2009;113:2508–2516. doi: 10.1182/blood-2008-05-158618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Hatton O, Phillips LK, Vaysberg M, Hurwich J, Krams SM, Martinez OM. Syk activation of phosphatidylinositol 3-kinase/Akt prevents HtrA2-dependent loss of X-linked inhibitor of apoptosis protein (XIAP) to promote survival of Epstein-Barr virus+ (EBV+) B cell lymphomas. J Biol Chem. 2011;286:37368–37378. doi: 10.1074/jbc.M111.255125. LMP-2 signaling through the Syk and the PI3K/Akt pathway in EBV-positive lymphomas.
  • 37.Chen L, Monti S, Juszczynski P, et al. SYK-dependent tonic B-cell receptor signaling is a rational treatment target in diffuse large B-cell lymphoma. Blood. 2008;111:2230–2237. doi: 10.1182/blood-2007-07-100115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Gururajan M, Dasu T, Shahidain S, et al. Spleen tyrosine kinase (Syk), a novel target of curcumin, is required for B lymphoma growth. J Immunol. 2007;178:111–121. doi: 10.4049/jimmunol.178.1.111. [DOI] [PubMed] [Google Scholar]
  • 39.Suljagic M, Longo PG, Bennardo S, et al. The Syk inhibitor fostamatinib disodium (R788) inhibits tumor growth in the Emu-TCL1 transgenic mouse model of CLL by blocking antigen-dependent B-cell receptor signaling. Blood. 2010;116:4894–4905. doi: 10.1182/blood-2010-03-275180. [DOI] [PubMed] [Google Scholar]
  • 40.Friedberg JW, Sharman J, Sweetenham J, et al. Inhibition of Syk with fostamatinib disodium has significant clinical activity in non-Hodgkin lymphoma and chronic lymphocytic leukemia. Blood. 2010;115:2578–2585. doi: 10.1182/blood-2009-08-236471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Braselmann S, Taylor V, Zhao H, et al. R406, an orally available spleen tyrosine kinase inhibitor blocks fc receptor signaling and reduces immune complex-mediated inflammation. J Pharmacol Exp Ther. 2006;319:998–1008. doi: 10.1124/jpet.106.109058. [DOI] [PubMed] [Google Scholar]
  • 42.Caldwell RG, Brown RC, Longnecker R. Epstein-Barr virus LMP2A-induced B-cell survival in two unique classes of EmuLMP2A transgenic mice. J Virol. 2000;74:1101–1113. doi: 10.1128/jvi.74.3.1101-1113.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bultema R, Longnecker R, Swanson-Mungerson M. Epstein-Barr virus LMP2A accelerates MYC-induced lymphomagenesis. Oncogene. 2009;28:1471–1476. doi: 10.1038/onc.2008.492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Rovedo M, Longnecker R. Epstein-Barr virus latent membrane protein 2A preferentially signals through the Src family kinase Lyn. J Virol. 2008;82:8520–8528. doi: 10.1128/JVI.00843-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Dargart JL, Fish K, Gordon LI, Longnecker R, Cen O. Dasatinib therapy results in decreased B cell proliferation, splenomegaly, and tumor growth in a murine model of lymphoma expressing Myc and Epstein-Barr virus LMP2A. Antiviral Res. 2012;95:49–56. doi: 10.1016/j.antiviral.2012.05.003. Dasatinib inhibts LMP-2-mediated activation of the Lyn pathway and leads to regression of EBV(+) lymphomas in a murine model.
  • 46.Dawson CW, Tramountanis G, Eliopoulos AG, Young LS. Epstein-Barr virus latent membrane protein 1 (LMP1) activates the phosphatidylinositol 3-kinase/Akt pathway to promote cell survival and induce actin filament remodeling. J Biol Chem. 2003;278:3694–3704. doi: 10.1074/jbc.M209840200. [DOI] [PubMed] [Google Scholar]
  • 47.Lambert SL, Martinez OM. Latent membrane protein 1 of EBV activates phosphatidylinositol 3-kinase to induce production of IL-10. J Immunol. 2007;179:8225–8234. doi: 10.4049/jimmunol.179.12.8225. [DOI] [PubMed] [Google Scholar]
  • 48. Hatton O, Lambert SL, Krams SM, Martinez OM. Src kinase and Syk activation initiate PI3K signaling by a chimeric latent membrane protein 1 in Epstein-Barr virus (EBV)+ B cell lymphomas. PLoS One. 2012;7:e42610. doi: 10.1371/journal.pone.0042610. LMP-1 activates the P13K/Akt pathway through Syk activation.
  • 49.Meckes DG, Jr, Menaker NF, Raab-Traub N. EBV LMP1 Modulates Lipid Raft Microdomains and the Vimentin Cytoskeleton for Signal Transduction and Transformation. J Virol. 2012 doi: 10.1128/JVI.02519-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kanemitsu N, Isobe Y, Masuda A, et al. Expression of Epstein-Barr virus-encoded proteins in extranodal NK/T-cell Lymphoma, nasal type (ENKL): differences in biologic and clinical behaviors of LMP1-positive and -negative ENKL. Clin Cancer Res. 2012;18:2164–2172. doi: 10.1158/1078-0432.CCR-11-2395. [DOI] [PubMed] [Google Scholar]
  • 51.Or YY, Hui AB, To KF, Lam CN, Lo KW. PIK3CA mutations in nasopharyngeal carcinoma. Int J Cancer. 2006;118:1065–1067. doi: 10.1002/ijc.21444. [DOI] [PubMed] [Google Scholar]
  • 52.Ma BB, Lui VW, Hui EP, et al. The activity of mTOR inhibitor RAD001 (everolimus) in nasopharyngeal carcinoma and cisplatin-resistant cell lines. Invest New Drugs. 2010;28:413–420. doi: 10.1007/s10637-009-9269-x. [DOI] [PubMed] [Google Scholar]
  • 53.Mei YP, Zhou JM, Wang Y, et al. Silencing of LMP1 induces cell cycle arrest and enhances chemosensitivity through inhibition of AKT signaling pathway in EBV-positive nasopharyngeal carcinoma cells. Cell Cycle. 2007;6:1379–1385. doi: 10.4161/cc.6.11.4274. [DOI] [PubMed] [Google Scholar]
  • 54.Ma BB, Lui VW, Hui CW, et al. Preclinical evaluation of the AKT inhibitor MK-2206 in nasopharyngeal carcinoma cell lines. Invest New Drugs. 2012 doi: 10.1007/s10637-012-9896-5. [DOI] [PubMed] [Google Scholar]
  • 55. Green MR, Rodig S, Juszczynski P, et al. Constitutive AP-1 activity and EBV infection induce PD-L1 in Hodgkin lymphomas and posttransplant lymphoproliferative disorders: implications for targeted therapy. Clin Cancer Res. 2012;18:1611–1618. doi: 10.1158/1078-0432.CCR-11-1942. EBV as a mechanism of PD-L1 induction in EBV-positive lymphomas.
  • 56. Shirley CM, Chen J, Shamay M, et al. Bortezomib induction of C/EBPbeta mediates Epstein-Barr virus lytic activation in Burkitt lymphoma. Blood. 2011;117:6297–6303. doi: 10.1182/blood-2011-01-332379. Bortezomib induces the unfolded protein response and CEBP-α-mediated lytic activation of EBV in Burkitt lymphoma.
  • 57.Sides MD, Block GJ, Shan B, et al. Arsenic mediated disruption of promyelocytic leukemia protein nuclear bodies induces ganciclovir susceptibility in Epstein-Barr positive epithelial cells. Virology. 2011;416:86–97. doi: 10.1016/j.virol.2011.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Sivachandran N, Wang X, Frappier L. Functions of the Epstein-Barr virus EBNA1 protein in viral reactivation and lytic infection. J Virol. 2012;86:6146–6158. doi: 10.1128/JVI.00013-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Li R, Wang L, Liao G, et al. SUMO binding by the Epstein-Barr virus protein kinase BGLF4 is crucial for BGLF4 function. J Virol. 2012;86:5412–5421. doi: 10.1128/JVI.00314-12. Role of SUMO binding of BGLF4 in the DNA damage response and EBV replication.
  • 60.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: 10.1002/ijc.24945. [DOI] [PubMed] [Google Scholar]
  • 61. Ghosh SK, Perrine SP, Williams RM, Faller DV. Histone deacetylase inhibitors are potent inducers of gene expression in latent EBV and sensitize lymphoma cells to nucleoside antiviral agents. Blood. 2012;119:1008–1017. doi: 10.1182/blood-2011-06-362434. Lytic induction of EBV with histone deacetylase inhibitors.
  • 62. Wildeman MA, Novalic Z, Verkuijlen SA, et al. Cytolytic virus activation therapy for epstein-barr virus-driven tumors. Clin Cancer Res. 2012;18:5061–5070. doi: 10.1158/1078-0432.CCR-12-0574. Lytic induction of EBV with gemcitabine and valproic acid.
  • 63.Jones K, Nourse J, Corbett G, Gandhi MK. Sodium valproate in combination with ganciclovir induces lysis of EBV-infected lymphoma cells without impairing EBV-specific T-cell immunity. Int J Lab Hematol. 2010;32:e169–e174. doi: 10.1111/j.1751-553X.2008.01130.x. [DOI] [PubMed] [Google Scholar]
  • 64.Perrine SP, Hermine O, Small T, 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: 10.1182/blood-2006-01-024703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Jung EJ, Lee YM, Lee BL, Chang MS, Kim WH. Lytic induction and apoptosis of Epstein-Barr virus-associated gastric cancer cell line with epigenetic modifiers and ganciclovir. Cancer Lett. 2007;247:77–83. doi: 10.1016/j.canlet.2006.03.022. [DOI] [PubMed] [Google Scholar]
  • 66. Li Y, Zhang Y, Fu M, et al. Parthenolide induces apoptosis and lytic cytotoxicity in Epstein-Barr virus-positive Burkitt lymphoma. Mol Med Report. 2012;6:477–482. doi: 10.3892/mmr.2012.959. Lytic induction of EBV with parthenolide.
  • 67. Hagemeier SR, Barlow EA, Meng Q, Kenney SC. The cellular ataxia telangiectasia-mutated (ATM) kinase promotes Epstein-Barr virus (EBV) lytic reactivation in response to multiple different types of lytic-inducing stimuli. J Virol. 2012 doi: 10.1128/JVI.01850-12. Role of ATM kinase in EBV lytic activation.
  • 68.Sokal EM, Hoppenbrouwers K, Vandermeulen C, et al. 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;196:1749–1753. doi: 10.1086/523813. [DOI] [PubMed] [Google Scholar]
  • 69.Moutschen M, Leonard P, Sokal EM, et al. Phase I/II studies to evaluate safety and immunogenicity of a recombinant gp350 Epstein-Barr virus vaccine in healthy adults. Vaccine. 2007;25:4697–4705. doi: 10.1016/j.vaccine.2007.04.008. [DOI] [PubMed] [Google Scholar]
  • 70.Elliott SL, Suhrbier A, Miles JJ, et al. Phase I trial of a CD8+ T-cell peptide epitope-based vaccine for infectious mononucleosis. J Virol. 2008;82:1448–1457. doi: 10.1128/JVI.01409-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Cohen JI, Fauci AS, Varmus H, Nabel GJ. Epstein-Barr virus: an important vaccine target for cancer prevention. Sci Transl Med. 2011;3:107fs107. doi: 10.1126/scitranslmed.3002878. Overview of EBV vaccine development, clinical trials, and future directions.
  • 72.Rees L, Tizard EJ, Morgan AJ, et al. A phase I trial of epstein-barr virus gp350 vaccine for children with chronic kidney disease awaiting transplantation. Transplantation. 2009;88:1025–1029. doi: 10.1097/TP.0b013e3181b9d918. [DOI] [PubMed] [Google Scholar]

RESOURCES