The Epstein–Barr virus (EBV) is an oncogenic, γ-herpesvirus associated with a broad spectrum of disease. Although most immune-competent individuals can effectivley develop efficient adaptive immune responses to EBV, immunocompromised individuals are at serious risk for developing life-threatening diseases, such as Hodgkin lymphoma and posttransplant lymphoproliferative disorder (PTLD). Given the significant morbidity associated with EBV infection in high-risk populations, there is a need to develop vaccine strategies that restore or enhance EBV-specific immune responses. Here, we identify the EBV immediate-early protein BZLF1 as a potential target antigen for vaccine development. Primary tumors from patients with PTLD and a chimeric human-murine model of EBV-driven lymphoproliferative disorder (EBV-LPD) express BZLF1 protein. Pulsing human dendritic cells (DC) with recombinant BZLF1 followed by incubation with autologous mononuclear cells led to expansion of BZLF1-specific CD8+ T cells in vitro and primed BZLF1-specific T-cell responses in vivo. In addition, vaccination of hu-PBL-SCID mice with BZLF1-transduced DCs induced specific cellular immunity and significantly prolonged survival from fatal EBV-LPD. These findings identify BZLF1 as a candidate target protein in the immunosurveillance of EBV and provide a rationale for considering BZLF1 in vaccine strategies to enhance primary and recall immune responses and potentially prevent EBV-associated diseases. Cancer Immunol Res; 3(7); 787–94. ©2015 AACR.

The Epstein–Barr virus (EBV) is a B-cell–lymphotropic, γ-herpesvirus that infects greater than 90% of the world's population. EBV causes infectious mononucleosis and has a clear association with several lymphoid and epithelial-derived malignancies, including Burkitt lymphomas, Hodgkin disease, and gastric and nasopharyngeal carcinomas (1). Following primary infection, the virus establishes lifelong persistance within the host memory B-cell compartment utilizing restricted latent gene expression programs that can manifest as either resting state (Lat I) or cellular immortalization and transformation programs (Lat II, III; refs. 2–5). In immune-competent hosts, the outgrowth of EBV-immortalized B cells is prevented through a robust expansion of EBV-specific memory T cells directed toward both lytic and latent antigens (6). EBV-positive individuals that possess either congenital or acquired immune deficiencies are thus highly susceptible to viral reactivation and malignant transformation. Furthermore, EBV-seropositive patients treated with immune suppressive medications following organ/stem-cell transplantation are at significant risk for developing posttransplant lymphoproliferative disorder (PTLD), an often fatal B-cell lymphoproliferatve disease (LPD; ref. 7).

Current treatment options for these patients are limited, and although chemo/immunotherapeutic reagents can induce tumor remission, these therapies often lead to opportunistic infections and short disease-free survival due to ablation of EBV-specific immunity (8). Thus, new therapeutic and preventive approaches are needed that specifically target EBV-infected cells while promoting long-term, protective immunity. The development of adoptive cellular therapeutic strategies has proven to be effective (9–11); however, these options have limited availability, are not yet universally applicable, and may be impractical for preventing EBV-associated diseases in solid organ transplant patients. Alternatively, the development of vaccination strategies to enhance primary and recall memory immune responses is receiving increasing consideration as both therapeutic and preventive approaches to EBV-associated diseases, including infectious mononucleosis (12–15). However, progress in this area has been markedly slow, partly because biomarkers that correlate with host protection are not fully known.

The immediate-early protein BZLF1 is a transcriptional activator that initiates the disruption of latency in EBV-infected cells. Upon expression, BZLF1 binds to the promoters of early EBV lytic genes, triggering a signaling cascade that results in viral DNA replication and virion production (16–19). BZLF1 has also been shown to inhibit antiviral cytokine signaling (20, 21), disrupt CD4+ T-cell recognition of MHC-II molecules (22), and promote B-cell transformation and lymphomagenesis in a humanized NOD/LtSz-scid/IL2Rγnull mouse model of EBV-related disease (23). Thus, BZLF1 expression is critical for viral activation, persistance, and disease pathogenesis, and, from an immunologic standpoint, may represent a prime target of adaptive immune responses. Indeed, BZLF1 is highly immunogenic and elicits robust CD4+ and CD8+ T-cell responses (6, 24, 25) that dominate the early immune responses in infectious mononucleosis patients and produce a healthy pool of EBV-specific memory T cells (26). Furthermore, we have shown that prevention of EBV-LPD in vivo is associated with the expansion of CD3+CD8+ T cells specific for BZLF1 (27). To the best of our knowledge, the potential therapeutic and preventative effects of BZLF1 vaccination have not been explored.

Here, we show that BZLF1 is expressed in EBV-LPD and is capable of eliciting a robust, memory T-cell response in healthy individuals. Furthermore, we demonstrate that presentation of full-length BZLF1 by human dendritic cells (DC) promotes the expansion of antigen-specific T-cell immunity and prolongs survival in a humanized, spontaneous murine model of EBV-LPD. These data suggest that BZLF1 is a critical target protein triggering immunosurveillance of EBV-infected cells and should be considered as a candidate for strategies to develop an EBV vaccine.

Examination of EBV-LPD tumor expression

Deidentified, pathologist-verified EBV-LPD tumor samples were obtained from The Ohio State University Pathology Core Facility (Columbus, OH). Patient and hu-PBL-SCID EBV-LPD tumors were stained with anti-BZLF1 (1:300; Santa Cruz Biotechnology) and visualized by immunohistochemistry (IHC) according to Institutional Review Board (IRB)-approved protocol.

ELISPOT assays

Secretion of human IFNγ was detected with the Human IFNγ enzyme-linked immunosorbent spot (ELISPOT) Kit (Mabtech) using MultiScreen-HA plates (Millipore) as per the manufacturer's instructions. Spots were visualized and counted using an Immunospot Imaging Analyzer (Cellular Technology Ltd.). Spot-forming units were calculated as the ratio between spots formed in response to antigen stimulation and spots formed in response to anti-CD3 mAb stimulation.

Mice

Six-week-old C.B.-17 SCID mice were purchased from Taconic Farms and housed in a specific pathogen-free environment. Animals showed no evidence of leaky phenotype (28) as determined by murine IgG ELISA (29). Animal work was approved by The Ohio State University Institutional Animal Care and Use Committee.

Flow cytometric analyses

mAbs specific for the following antigens were used: anti-human CD3, CD8, and IFNγ (BD Bioscience). Human HLA-B8 tetramers complexed with allele immunodominant peptides for BZLF1 (RAKFKQLL; RAK) and EBNA-3A (FLRGRAYGL; FLR) and conjugated with allophycocyanin (APC) were provided by the NIAID Tetramer Facility and the NIH AIDS Research and Reference Reagent Program (27). Events were collected on an FC500 flow cytometer (Beckman-Coulter) and analyzed with FlowJo v7.6.1 (TreeStar) software.

Lymphoblastoid cell line generation and coculture

EBV-transformed lymphoblastoid cell lines (LCL) were derived in vivo by engrafting SCID mice with human peripheral blood mononuclear cells (PBMC) from healthy EBV-positive donors as described (30). Cocultures for ELISPOT and IFNγ flow assays were created in 96-well U-bottom plates by combining irradiated LCLs (14,000 rad) with equal numbers of autologous PBMCs. Cultures were maintained in RPMI-1640 containing 10% FBS supplemented with antibiotics and 1% Glutamax (Life Technologies) in the presence of 10 U/mL IL2 (Prometheus).

Cloning, expression, and purification of recombinant (r) BZLF1

The open reading frame sequence of EBV BZLF1 was amplified by RT-PCR using RNA isolated from an EBV-positive LCLs; forward primer: ATGATGGACCCAAACT, reverse primer: TTAGAAATTTAAGAG. The amplified product was cloned into Nde I/Not I sites of the pET26b(+) vector (Novagen), and subsequently introduced into Escherichia coli strain BL26 (Invitrogen) by standard transformation. Expression of BZLF1 protein was induced by 1.5 nmol/L IPTG treatment for 3 hours. In collaboration with ARVYS Proteins Inc., milligram quantities of low endotoxin-rBZLF1 protein (GMP-grade materials) were purified by a method comprising recovery and washing of inclusion bodies, two gel filtration chromatography steps, and filtration through anion-exchange filters. The identity of rBZLF1 protein was confirmed by mass spectrometry (Nano-LC/MS/MS system).

Generation of rAd5F35/BZLF1 virus

The rAd5F35/BZLF1 adenovirus was generated using the Adeno-X expression system 1 (Clontech Laboratories, Inc.). In brief, BZLF1 cDNA was cloned into Not I sites of the mammalian expression cassette pShuttle2. The I-CeuI/PI-SceI fragment containing the BZLF1 expression cassette was gel-purified and subcloned into I-CeuI/PI-SceI sites of the AdF35 genome (a gift from Dr. Malcolm Brenner, the Baylor College of Medicine, Houston, TX), in which the fiber gene of adenovirus serotype 35 is incorporated into an Ad5 capsid. Finally, Pac-I–digested pAd5F35/BZLF1 DNA was used to transfect HEK 293 cells. The individual viral plaque was isolated, and positive viral clones were used for large-scale production of rAd5F35/BZLF1 stock. The viral titer was determined using the Adeno-X rapid titer Kit (Clontech Laboratories, Inc.).

Generation of rAAV2/BZLF1 virus

The rAAV2/BZLF1 virus consists of rAAV terminal repeats flanking a CMV-IE promoter BZLF1 gene expression cassette. To produce the rAAV2/BZLF1 vector, a Hela-derived cell line containing the above sequences, the AAV rep and cap genes, and a neomycin-resistant selectable marker was constructed as described (31). The rAAV2/GFP virus is identical to rAAV2/BZLF1 except the GFP gene is driven by the CMV-IE promoter. Large-scale rAAV2 viral preparation, purification, and quantification were performed as described (32–34).

In vitro culture and infection of 293T cells

The human amphotropic-packaging cell line HEK 293T (ATCC) was maintained in a 37°C humidified environment supplemented with 5% CO2 in DMEM media containing 5% FBS and grown to 80% confluence before infection. Following incubation, cells were cultured in the presence of rAAV2/BZLF1 [multiplicity of infection (MOI) = 10], rAAV2/GFP (MOI = 10), rAd5F35/BZLF1 (MOI = 10), or rAd5F35/null (MOI = 10) for 3 hours.

Immunoblotting

All immunoblotting was performed as described (35), using antibodies directed against BZLF1 (Santa Cruz Biotechnology) and actin (Santa Cruz Biotechnology). Staining of SDS-PAGE gels was performed using Coomassie Brilliant Blue Staining Solution (Bio-Rad).

In vitro DC generation and culture

PBMCs were obtained from healthy donors under an OSU IRB-approved protocol. Following Ficoll–Hypaque (GE healthcare Bioscience AB) separation and incubation of 5 × 106 cells/mL in serum-free media for 2 hours at 37°C, the supernatant containing nonadherent cells was removed. Adherent monocytes were cultured for 3 days with human GM-CSF (50 ng/mL; Peprotech) and IFNα (1,000 U/mL; Schering Corporation) in the presence of rBZLF1 protein or an equivalent amount of BSA (control), or rAd5F35/BZLF1 (MOI = 10) or rAd5F35/null (MOI = 10; Vector Biolabs), or rAAV2/BZLF1 or rAAV2/GFP, respectively (36). DCs were then cocultured with autologous HLA-B8+ PBMCs at a ratio of 1:5 in the presence of 10 IU/mL IL2 for 10 days.

In vivo studies

The engrafted humanized SCID (hu-PBL-SCID) mouse model of EBV-LPD has been described (37). Six-week-old SCID mice were injected i.p. with 5 × 107 PBMCs that were obtained from a healthy HLA-B8+ EBV-seropositive donor. For depletion of endogenous murine natural killer (NK) cells, SCID mice were injected i.p. with anti–asialo-ganglioside 1 antiserum (ASGM-1; Wako Chemicals) 1 day before injection of human PBMCs and every 7 days thereafter for the duration of the study. Engraftment was confirmed by human IgG ELISA as described (37). Intraperitoneal vaccination with DCs previously cocultured with rBZLF1 or control BSA or virally transduced DCs (described above) began immediately following initial engraftment.

Statistical analyses

For the comparisons of IFNγ secretion in Figs. 2, 3E, and 5A, two sample t tests were used. Log-rank tests were used for the survival analyses in Fig. 5, and Kaplan–Meier survival curves were generated to display the results. All analyses were performed using SAS/STAT software version 9.2 (SAS Institute Inc.).

BZLF1 is expressed in human EBV-LPD

Recognition of EBV-encoded epitopes by effector lymphocytes is a major component of the adaptive immune system's ability to clear lytic and latently infected B cells. To determine whether BZLF1 is expressed in EBV+ malignancies, we performed IHC assays on primary EBV+ lymphoma tumor samples using an antibody specific for BZLF1. Of the tumors analyzed, 6 of 8 PTLD samples stained positive for BZLF1 (Fig. 1A and Table 1). Other EBV+ lymphomas staining for BZLF1 included lymphomatoid granulomatosis (1/3), diffuse large B-cell lymphoma (5/10), and double-hit lymphoma (1/1). Both EBV+ Hodgkin lymphoma (6 cases) and EBV+ unclassifiable lymphomas (8 cases) failed to demonstrate any BZLF1 signal. We next determined whether BZLF1 is expressed in the EBV-LPD tumors that spontaneously developed in hu-PBL-SCID mice engrafted from 3 separate donors of various HLA types (30). Consistent with our patient data, all hu-PBL-SCID tumors were positive for the BZLF1 protein (Fig. 1B). These results led us to hypothesize that enhanced recognition of BZLF1 by antigen-specific cellular immunity may provide a protective effect against the development of fatal EBV-LPD in hu-PBL-SCID mice.

Figure 1.

BZLF1 gene expression in human EBV+ lymphoma samples. A, immunohistochemical staining of a pathologist-verified EBV+ PTLD patient sample with anti-BZLF1. The brown staining indicates the presence of BZLF1 protein; left, ×20 magnification; right, ×40 magnification. The images are representative of 6 patient samples. B, immunohistochemical staining of EBV-LPD tumor sample obtained from hu-PBL-SCID mouse. Brown staining indicates presence of BZLF1 protein; left, ×20 magnification; right, ×40 magnification. Data are representative of 5 tumor-bearing hu-PBL-SCID mice.

Figure 1.

BZLF1 gene expression in human EBV+ lymphoma samples. A, immunohistochemical staining of a pathologist-verified EBV+ PTLD patient sample with anti-BZLF1. The brown staining indicates the presence of BZLF1 protein; left, ×20 magnification; right, ×40 magnification. The images are representative of 6 patient samples. B, immunohistochemical staining of EBV-LPD tumor sample obtained from hu-PBL-SCID mouse. Brown staining indicates presence of BZLF1 protein; left, ×20 magnification; right, ×40 magnification. Data are representative of 5 tumor-bearing hu-PBL-SCID mice.

Close modal
Table 1.

BZLF1 expression in EBV+ primary

EBV+ lymphomasBZLF1(+)a% Tumor BZLF1(+)b
PTLD 6/8 (75%) 1%–10% 
Lymphomatoid granulomatosis 1/3 (33%) 1% 
Diffuse large B-cell lymphoma 5/10 (50%) 1%–10% 
Hodgkin lymphoma 0/6 (0%) 0% 
EBV+ unclassifiable lymphoma 0/8 (0%) 0% 
EBV+ lymphomasBZLF1(+)a% Tumor BZLF1(+)b
PTLD 6/8 (75%) 1%–10% 
Lymphomatoid granulomatosis 1/3 (33%) 1% 
Diffuse large B-cell lymphoma 5/10 (50%) 1%–10% 
Hodgkin lymphoma 0/6 (0%) 0% 
EBV+ unclassifiable lymphoma 0/8 (0%) 0% 

aPositive expression for BZLF1 was documented for any tumor that demonstrated anti-BZLF1 signal by immunohistochemical analysis.

bPercentage of tumor cellularity that is BZLF1(+).

BZLF1 recognition triggers a cell-mediated immune response in healthy carriers

We next evaluated the cell-mediated immune responses against BZLF1 in vitro. Antigen recognition by the adaptive immune system, specifically by T-cell subsets, is essential to providing long-term, protective immunity against EBV (6). PBMCs were isolated from 24 healthy, EBV-seropositive donors of various HLA types and infected with adenovirus encoding a recombinant BZLF1 (rAd5F35/BZLF1) or null control (rAd5F35/null) for 18 hours without cytokine treatment. To quantify the immune responses against BZLF1, an IFNγ ELISPOT assay was used to detect the number of cytokine-secreting T cells. The degree of responsiveness, as determined by level of IFNγ secretion, varied with each donor, highlighting the inherent diversity of antigen exposure and recall responses (Fig. 2).

Figure 2.

Donor immune responses to BZLF1 antigen exposure. PBMCs from 24 donors of various HLA types were infected with adenovirus carrying either BZLF1 cDNA (rAd5F35/BZLF1) or an empty-vector control (rAd5F35/null) for 18 hours without cytokine treatment. Human IFNγ ELISPOT assays were performed immediately following infection with virus. Spot-forming units were calculated as described above. Bars show mean ± SD. *, P ≤ 0.05 as determined by two-sample t test.

Figure 2.

Donor immune responses to BZLF1 antigen exposure. PBMCs from 24 donors of various HLA types were infected with adenovirus carrying either BZLF1 cDNA (rAd5F35/BZLF1) or an empty-vector control (rAd5F35/null) for 18 hours without cytokine treatment. Human IFNγ ELISPOT assays were performed immediately following infection with virus. Spot-forming units were calculated as described above. Bars show mean ± SD. *, P ≤ 0.05 as determined by two-sample t test.

Close modal

Recombinant BZLF1 vaccination promotes antigen-specific T-cell expansion and function in vitro and in vivo

We next examined whether DCs loaded with BZLF1 protein could mediate the expansion of BZLF1-specific memory CD8+ T cells in vitro. Milligram quantities of rBZLF1 protein were generated as described above and analyzed for purity by SDS-PAGE analysis (Fig. 3A). Purified rBZLF1 was then loaded onto monocyte-derived DCs at varying concentrations and cocultured with autologous PBMCs from an HLA-B8+ donor for 10 days (Fig. 3B). To analyze the ability of rBZLF1 protein to generate BZLF1-specific CTLs, a flow cytometric tetramer assay was used to detect the immunodominant HLA-B8–restricted RAK epitope of the BZLF1 protein (27). BZLF1-presenting DCs generated RAK-specific CD8+ T cells in a dose-dependent fashion (Fig. 3C). To assess the functional status of these T-cell subsets, we restimulated the DC-PBMC cocultures at day 13 with autologous LCLs. Restimulation induced a RAK-specific IFNγ response that was mediated by CD8+ subsets (Fig. 3D). Thus, BZLF1 antigen exposure by DCs confers specific CD8+ T-cell immunity in vitro for PBMCs from an HLA-B8+ individual.

Figure 3.

Recombinant BZLF1 presentation by human DCs promotes BZLF1-specific CD8+ T-cell expansion and function. A, SDS-PAGE protein gel stained with Coomassie Brilliant Blue. Gel contains low-range molecular weight markers and rBZLF1 protein at the indicated quantities. B, schematic representation of experimental approach. C, flow cytometric RAK HLA-B8 tetramer assay of PBMC cocultures expanded with rBZLF1-loaded DCs. D, IFNγ intracellular flow cytometric analysis of DC-expanded PBMC cocultures restimulated with autologous LCLs for 18 hours. An HLA-B8–restricted FLR tetramer derived from the EBV latency protein EBNA-3A was used as a negative control. E, IFNγ ELISPOT assay of ex vivo splenocytes collected from hu-PBL-SCID mice vaccinated with rBZLF1 protein and restimulated with BZLF1-loaded DCs in vitro (P = 0.0007, n = 3). *, P ≤ 0.05 as determined by two-sample t test.

Figure 3.

Recombinant BZLF1 presentation by human DCs promotes BZLF1-specific CD8+ T-cell expansion and function. A, SDS-PAGE protein gel stained with Coomassie Brilliant Blue. Gel contains low-range molecular weight markers and rBZLF1 protein at the indicated quantities. B, schematic representation of experimental approach. C, flow cytometric RAK HLA-B8 tetramer assay of PBMC cocultures expanded with rBZLF1-loaded DCs. D, IFNγ intracellular flow cytometric analysis of DC-expanded PBMC cocultures restimulated with autologous LCLs for 18 hours. An HLA-B8–restricted FLR tetramer derived from the EBV latency protein EBNA-3A was used as a negative control. E, IFNγ ELISPOT assay of ex vivo splenocytes collected from hu-PBL-SCID mice vaccinated with rBZLF1 protein and restimulated with BZLF1-loaded DCs in vitro (P = 0.0007, n = 3). *, P ≤ 0.05 as determined by two-sample t test.

Close modal

To determine whether vaccination with rBZLF1 could prime BZLF1-specific T-cell immunity in vivo, hu-PBL-SCID mice were generated using HLA-B8+ PBMCs from a single EBV+ donor and injected once with either PBS, GM-CSF (5 μg/mouse), rBZLF1 (100 μg/mouse), or rBZLF1 with GM-CSF as an immune adjuvant (27). Four weeks after vaccination, ex vivo splenocytes were reexposed to rBZLF1-presenting DCs in vitro and assayed for IFNγ secretion by ELISPOT. In comparison with vehicle, vaccination with rBZLF1 alone significantly enhanced BZLF1-specific IFNγ responses (P = 0.0007, n = 3, Fig. 3E), indicating an efficient expansion of functional, BZLF1-specific T cells. Coadministration of rBZLF1 and GM-CSF did not significantly enhance the effect to BZLF1 re-exposure.

BZLF1 virally transduced DCs mediate the expansion of BZLF1-specific CTLs in vitro

We next investigated if viral-mediated delivery of BZLF1 to DCs could serve as an effective vaccine strategy against EBV. We compared the ability of adenoviral and adeno-associated viral gene transfer to generate functional, BZLF1-presenting DCs in vitro. Monocyte-derived DCs were infected with either an adeno-associated virus (rAAV2/BZLF1) or adenovirus (rAdv5F35/BZLF1) encoding recombinant BZLF1 (or control); infection and overexpression of BZLF1 were confirmed in HEK 293T cells (Fig. 4A and C, respectively). Infected DCs were then cocultured with autologous HLA-B8+ PBMCs for 7 days and analyzed for RAK-specific CD8+ T-cell expansion by flow cytometry. DCs transduced with rAAV2/BZLF1 had a minimal effect on RAK-specific CTL expansion compared with DCs incubated with naked rBZLF1 protein (Fig. 4B); however, rAd5F35/BZLF1-transduced DCs induced a robust expansion of RAK-specific CD8+ T cells (Fig. 4D). Importantly, DCs transduced with rAd5F35/BZLF1 showed a greater expansion of antigen-specific T cells compared with that by DCs transduced with rAd5F35-null control or by rBZLF1-presenting DCs (compare Fig. 4B and D).

Figure 4.

BZLF1 virally transduced DCs induce BZLF1-specific T-cell immunity in vitro. A, immunoblot of BZLF1 protein expression in HEK 293T cells (lysate from 1 × 106 cells/lane) infected with adeno-associated virus encoding GFP alone (rAAV2/GFP) or GFP and BZLF1 (rAAV/BZLF1). B, flow cytometric HLA-B8 RAK tetramer assay of PBMC cocultures expanded with rAAV/GFP or rAAV/BZLF1-infected DCs; 1 μg/mL rBZLF1-loaded DCs were used as a positive control. C, immunoblot of BZLF1 protein expression in HEK 293T cells infected with rAd5F35/null or rAd5F35/BZLF1. D, flow cytometric HLA-B8 RAK tetramer assay of PBMC cocultures expanded with rAd5F35/null or rAd5F35/BZLF1-infected DCs. Nonreactive HLA-B8 FLR tetramer was used as a negative control.

Figure 4.

BZLF1 virally transduced DCs induce BZLF1-specific T-cell immunity in vitro. A, immunoblot of BZLF1 protein expression in HEK 293T cells (lysate from 1 × 106 cells/lane) infected with adeno-associated virus encoding GFP alone (rAAV2/GFP) or GFP and BZLF1 (rAAV/BZLF1). B, flow cytometric HLA-B8 RAK tetramer assay of PBMC cocultures expanded with rAAV/GFP or rAAV/BZLF1-infected DCs; 1 μg/mL rBZLF1-loaded DCs were used as a positive control. C, immunoblot of BZLF1 protein expression in HEK 293T cells infected with rAd5F35/null or rAd5F35/BZLF1. D, flow cytometric HLA-B8 RAK tetramer assay of PBMC cocultures expanded with rAd5F35/null or rAd5F35/BZLF1-infected DCs. Nonreactive HLA-B8 FLR tetramer was used as a negative control.

Close modal

In vivo vaccination of hu-PBL-SCID chimeric mice with rAd5F35/BZLF1-transduced DCs induces specific cellular immunity and delays EBV-LPD

The hu-PBL-SCID model of spontaneous EBV-LPD has been used to study potential experimental therapeutic strategies for EBV-LPD (37, 38). We examined whether vaccination of hu-PBL-SCID mice with rAd5F35/BZLF1-transduced DCs (or control) would positively enhance cellular immunity and improve survival against fatal EBV-LPD. HLA-B8+ PBMCs from a healthy EBV-seropositive donor and 5 × 106 autologous DCs transduced with either rAd5F35/BZLF1 or empty vector (rAd5F35/null) were injected i.p. into SCID mice (n = 10/group). Evidence of human cell engraftment was confirmed by assessing human IgG concentrations in serum. Two weeks after injection, mice in the “booster group” received a second i.p. injection of either rAd5F35/BZLF1-transduced or rAd5F35/null-transduced DCs as a booster. Five weeks after initial vaccination, 3 mice from the booster group were sacrificed and analyzed for immunosurveillance capacity against EBV-LPD. Splenocytes were cocultured ex vivo with autologous LCLs and assayed for IFNγ secretion by ELISPOT. Mice vaccinated with rAd5F35/BZLF1-transduced DCs showed significantly higher responsiveness to LCLs relative to that in vector-control–vaccinated mice (P < 0.0001, n = 3; Fig. 5A). Single vaccination with rAd5F35/BZLF1-transduced DCs showed a trend toward improved survival (P = 0.085, n = 10) at a later stage (Fig. 5B); however, vaccination followed by single booster delivery significantly delayed fatal EBV-LPD (P = 0.014, n = 10; Fig. 5C), with a median survival of 62 days for the rAd5F35/BZLF1 group and 48 days for the rAd5F35/null group.

Figure 5.

Evaluation of in vivo activity of BZLF1 virally transduced DC vaccination in hu-PBL-SCID mice. SCID mice depleted of murine NK cells were injected i.p. with 5 × 107 HLA-B8+EBV+ PBMCs from a single, healthy donor and 5 × 106 autologous monocyte-derived DCs transduced with either rAd5F35/null or rAd5F35/BZLF1. Two weeks after initial vaccination, mice in the “booster group” received a second i.p. injection with either rAd5F35/BZLF1 or rAd5F35/null transduced DCs (5 × 106 DCs per mouse). A, IFNγ ELISPOT assay of ex vivo splenocytes from booster group restimulated with autologous LCLs in vitro (P < 0.0001, n = 3). Bars show mean ± SD. B, Kaplan–Meier analysis of overall survival for mice that received a single dose vaccination (P = 0.085, n = 10 each). C, Kaplan–Meier analysis of overall survival for mice that received a single booster dose injection 2 weeks after primary vaccination (P = 0.014, n = 10 each). *, P ≤ 0.05 as determined by the log-rank analysis.

Figure 5.

Evaluation of in vivo activity of BZLF1 virally transduced DC vaccination in hu-PBL-SCID mice. SCID mice depleted of murine NK cells were injected i.p. with 5 × 107 HLA-B8+EBV+ PBMCs from a single, healthy donor and 5 × 106 autologous monocyte-derived DCs transduced with either rAd5F35/null or rAd5F35/BZLF1. Two weeks after initial vaccination, mice in the “booster group” received a second i.p. injection with either rAd5F35/BZLF1 or rAd5F35/null transduced DCs (5 × 106 DCs per mouse). A, IFNγ ELISPOT assay of ex vivo splenocytes from booster group restimulated with autologous LCLs in vitro (P < 0.0001, n = 3). Bars show mean ± SD. B, Kaplan–Meier analysis of overall survival for mice that received a single dose vaccination (P = 0.085, n = 10 each). C, Kaplan–Meier analysis of overall survival for mice that received a single booster dose injection 2 weeks after primary vaccination (P = 0.014, n = 10 each). *, P ≤ 0.05 as determined by the log-rank analysis.

Close modal

EBV is the etiological agent of infectious mononucleosis and one of few known human oncoviruses. Although most carriers possess the immune components needed to limit the outgrowth of latently infected B cells, immunocompromised patients, such as those with congenital or acquired immunodeficiencies or those having undergone organ transplantation, are at increased risk for malignant transformation due to diminished EBV-specific immunity. EBV is also associated with a broad spectrum of benign (multiple sclerosis, lupus) and malignant diseases (nasopharyngeal, gastric carcinomas) in various immune-competent populations; thus, strategies to enhance immunity to EBV represent an unmet need. In the current study, we demonstrate that vaccination of hu-PBL-SCID mice against the EBV immediate-early protein BZLF1 can enhance specific cellular immunity and significantly delay the development of lethal EBV-LPD. These protective effects are associated with the expansion and activation of CD8+ T-cell subsets that recognize the immunodominant HLA-B8–restricted RAK peptide of BZLF1.

Traditionally, EBV-associated malignancies are thought to be driven largely by the cooperative functions of latent viral gene products. Our data support the emerging hypothesis that BZLF1 gene expression is a prominent factor in the development of EBV-associated malignancy (23). We found that both patient and hu-PBL-SCID EBV-LPD tumors show BZLF1-expressing tumor cells that, when targeted in hu-PBL-SCID mice, delay the development of fatal EBV-LPD. Consistent with this observation, Ma and colleagues (23) recently demonstrated that humanized NOD/LtSz-scid/IL2Rγnull mice infected with a replication-defective BZLF1-deleted EBV mutant developed B-cell lymphomas less frequently than control virus–infected mice. Because BZLF1 expression has not currently been demonstrated to be sufficient to drive oncogenesis, its expression in malignant cells may provide a mechanism for evading tumor surveillance perhaps by downregulating proinflammatory cytokine signaling (20, 21) or by inhibiting the LMP1-induced upregulation of MHC class I molecules in infected cells (39). Horizontal EBV transmission to bystander EBV-negative B lymphocytes may also confer a distinct survival advantage to EBV+ lymphomas by replinishing latently infected B-cell populations (23).

Regardless, given the capacity for BZLF1 vaccination to promote EBV-specific immunity and, in turn, delay the progression of fatal EBV-LPD in hu-PBL-SCID mice, it is reasonable to hypothesize that immunosurveillance of BZLF1 by T-cell subsets is a critical factor in the pathogenesis of EBV-LPD and perhaps other associated diseases. Indeed, we have previously shown that prevention of EBV-LPD in hu-PBL-SCID mice treated with combination cytokine therapy is associated with a marked expansion of BZLF1-specific CD8+ lymphocytes (27). Furthermore, we have demonstrated that successful treatment of posttransplant LPD following renal allografting is associated with high frequencies of RAK-specific CD8+ T cells (40). Taken together, these findings point to the critical nature of BZLF1 immunosurveillance and the importance of restoring BZLF1-specific immunity in patients at risk or suffering from EBV-associated diseases.

Our vaccination experiment with BZLF1-transduced DCs clearly establishes that BZLF1 vaccination can induce specific cellular immunity capable of recognizing and eliminating EBV-transformed cells in hu-PBL-SCID mice. Indeed, the significant expansion of BZLF1-specific T cells (Figs. 3E and 5A), coupled with the increase in survival following repeated vaccination with BZLF1, suggests that BZLF1-specific T cells are expanding and clearing BZLF1-expressing tumor cells, and is doing so in a chimeric mouse model that is dominated by a dampened human immune response (41). Indeed, all vaccinated animals in our study ultimately developed fatal EBV-LPD. This effect is likely the result of hypofunctioning human T cells in the xenogeneic environment, the selection of viral antigen-loss mutants that have “hidden” BZLF1 from effector lymphocytes (42), and/or the presence of natural viral gene polymorphisms (43) that allow for immune escape. The latter possibility would support the rationale to develop a multivalent vaccine that includes additional target antigens covering a broad spectrum of EBV's genetic programs including proteins uniquely associated with malignancy. However, the single valent vaccine utilized in this study might be sufficient to prevent clinically fulminant infectious mononucleosis that can occur in genetically susceptible individuals (44). We are currently exploring multivalent vaccine approaches, including BZLF1, as one of the vaccine immunogens.

Although recombinant BZLF1 vaccination primed BZLF1-specific T-cell responses in vivo, it is particularly interesting that we did not observe an enhanced effect when GM-CSF was administered as an immune adjuvant (Fig. 3E), especially given our prior findings that combination GM-CSF and IL2 therapy protected hu-PBL-SCID mice from fatal EBV-LPD (27). Indeed, others have demonstrated that GM-CSF therapy enhances vaccine-mediated immune responses and confers protective immunity in vivo via the induction of professional APCs (45, 46). In contrast with these observations, however, Serafini and colleagues (47) have shown that high-dose GM-CSF therapy can impair antigen-specific T-cell expansion through the recruitment of myeloid-derived suppressor cells. Thus, in future vaccine studies with BZLF1, it may be useful to assess the delivery of low doses of immune adjuvants that promote specific cellular immunity while limiting the growth and recruitment of suppressor cell subsets.

Although our immunization strategy described here significantly prolonged survival from fatal EBV-LPD in hu-PBL-SCID mice engrafted with PBMCs from an HLA-B8+ individual, we cannot exclude the possibility that BZLF1 vaccination will not be as effective in individuals with other HLA types. Given that recognition of the immunodominant RAK peptide of BZLF1 is HLA-B8–restricted, it remains possible that BZLF1 exposure would produce a comparatively diminished immune response in individuals with other HLA types, especially if additional vaccine components compete with BZLF1 for T-cell receptor binding. Although this study does not directly compare cell-mediated immune responses to BZLF1 vaccination in HLA-B8 versus HLA-B8+ individuals, we did observe IFNγ production in donors with a variety of HLA tissue types (Fig. 2). Of note, HLA-B8 individuals tested in this experiment produced modest to low levels of IFNγ, suggesting that non–HLA-related mechanisms are responsible for mediating the immune response to this target protein. Future work will need to directly address distinct differences in EBV antigen responsiveness between individuals with various HLA types.

In summary, we have demonstrated that vaccination with the EBV immediate-early protein BZLF1, a trascription factor that initiates the transition to lytic productive program, can induce a protective effect against the development of fatal EBV-LPD in SCID mice engrafted with PBMCs from an HLA-B8+ individual. As the hu-PBL-SCID model described here appears to recapitulate the pathogenesis of human EBV-LPD, our findings support consideration of this vaccination strategy in patients, especially HLA-B8+ patients, who are at risk for EBV-LPD (48) and other EBV-associated diseases. Our findings also support the consideration of BZLF1 as a component in a multivalent vaccine that consists of multiple EBV gene products. Efforts to identify other latent and lytic antigens that can be included in the development of a multivalent vaccine are currently under way.

No potential conflicts of interest were disclosed.

Conception and design: T. Liu, X. Zhang, M.E. Lustberg, R.A. Baiocchi

Development of methodology: T. Liu, J.T. Patton, S.L. Garman, G. Lozanski, R.A. Baiocchi

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A.S. Hartlage, J.T. Patton, S.L. Garman, H. Kurt, G. Lozanski, R.A. Baiocchi

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A.S. Hartlage, T. Liu, S.L. Garman, X. Zhang, M.E. Lustberg, R.A. Baiocchi

Writing, review, and/or revision of the manuscript: A.S. Hartlage, T. Liu, J.T. Patton, S.L. Garman, X. Zhang, G. Lozanski, M.A. Caligiuri, R.A. Baiocchi

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S.L. Garman, R.A. Baiocchi

Study supervision: S.L. Garman, R.A. Baiocchi

This work was supported by The Friends of Jason Gould Foundation, the Leukemia Lymphoma Society Translational Research Program (to R.A. Baiocchi and M.A. Caligiuri), the National Institutes of Health (to R.A. Baiocchi and M.A. Caligiuri, 1RC2CA148348-01), and NIH grant T32CA090223.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Cohen
JI
. 
Epstein-Barr virus infection
.
N Engl J Med
2000
;
343
:
481
92
.
2.
Joseph
AM
,
Babcock
GJ
,
Thorley-Lawson
DA
. 
EBV persistence involves strict selection of latently infected B cells
.
J Immunol
2000
;
165
:
2975
81
.
3.
Babcock
GJ
,
Decker
LL
,
Volk
M
,
Thorley-Lawson
DA
. 
EBV persistence in memory B cells in vivo
.
Immunity
1998
;
9
:
395
404
.
4.
Miller
G
. 
Immortalization of human lymphocytes by Epstein-Barr virus
.
Yale J Biol Med
1982
;
55
:
305
10
.
5.
Rowe
M
,
Lear
AL
,
Croom-Carter
D
,
Davies
AH
,
Rickinson
AB
. 
Three pathways of Epstein-Barr virus gene activation from EBNA1-positive latency in B lymphocytes
.
J Virol
1992
;
66
:
122
31
.
6.
Tan
LC
,
Gudgeon
N
,
Annels
NE
,
Hansasuta
P
,
O'Callaghan
CA
,
Rowland-Jones
S
, et al
A re-evaluation of the frequency of CD8+ T cells specific for EBV in healthy virus carriers
.
J Immunol
1999
;
162
:
1827
35
.
7.
Loren
AW
,
Porter
DL
,
Stadtmauer
EA
,
Tsai
DE
. 
Post-transplant lymphoproliferative disorder: a review
.
Bone Marrow Transplant
2003
;
31
:
145
55
.
8.
Heslop
HE
. 
How I treat EBV lymphoproliferation
.
Blood
2009
;
114
:
4002
8
.
9.
Louis
CU
,
Straathof
K
,
Bollard
CM
,
Ennamuri
S
,
Gerken
C
,
Lopez
TT
, et al
Adoptive transfer of EBV-specific T cells results in sustained clinical responses in patients with locoregional nasopharyngeal carcinoma
.
J Immunother
2010
;
33
:
983
90
.
10.
Comoli
P
,
De Palma
R
,
Siena
S
,
Nocera
A
,
Basso
S
,
Del Galdo
F
, 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
7
.
11.
Icheva
V
,
Kayser
S
,
Wolff
D
,
Tuve
S
,
Kyzirakos
C
,
Bethge
W
, 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
.
12.
Duraiswamy
J
,
Sherritt
M
,
Thomson
S
,
Tellam
J
,
Cooper
L
,
Connolly
G
, et al
Therapeutic LMP1 polyepitope vaccine for EBV-associated Hodgkin disease and nasopharyngeal carcinoma
.
Blood
2003
;
101
:
3150
6
.
13.
Lutzky
VP
,
Corban
M
,
Heslop
L
,
Morrison
LE
,
Crooks
P
,
Hall
DF
, et al
Novel approach to the formulation of an Epstein-Barr virus antigen-based nasopharyngeal carcinoma vaccine
.
J Virol
2010
;
84
:
407
17
.
14.
Ruiss
R
,
Jochum
S
,
Wanner
G
,
Reisbach
G
,
Hammerschmidt
W
,
Zeidler
R
. 
A virus-like particle-based Epstein-Barr virus vaccine
.
J Virol
2011
;
85
:
13105
13
.
15.
Sokal
EM
,
Hoppenbrouwers
K
,
Vandermeulen
C
,
Moutschen
M
,
Leonard
P
,
Moreels
A
, 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
53
.
16.
Rooney
CM
,
Rowe
DT
,
Ragot
T
,
Farrell
PJ
. 
The spliced BZLF1 gene of Epstein-Barr virus (EBV) transactivates an early EBV promoter and induces the virus productive cycle
.
J Virol
1989
;
63
:
3109
16
.
17.
Countryman
J
,
Jenson
H
,
Seibl
R
,
Wolf
H
,
Miller
G
. 
Polymorphic proteins encoded within BZLF1 of defective and standard Epstein-Barr viruses disrupt latency
.
J Virol
1987
;
61
:
3672
9
.
18.
Farrell
PJ
,
Rowe
DT
,
Rooney
CM
,
Kouzarides
T
. 
Epstein-Barr virus BZLF1 trans-activator specifically binds to a consensus Ap-1 site and is related to C-Fos
.
EMBO J
1989
;
8
:
127
32
.
19.
Fixman
ED
,
Hayward
GS
,
Hayward
SD
. 
Replication of Epstein-Barr-virus orilyt - lack of a dedicated virally encoded origin-binding protein and dependence on Zta in cotransfection assays
.
J Virol
1995
;
69
:
2998
3006
.
20.
Morrison
TE
,
Mauser
A
,
Klingelhutz
A
,
Kenney
SC
. 
Epstein-Barr virus immediate-early protein BZLF1 inhibits tumor necrosis factor alpha-induced signaling and apoptosis by downregulating tumor necrosis factor receptor 1
.
J Virol
2004
;
78
:
544
9
.
21.
Morrison
TE
,
Mauser
A
,
Wong
A
,
Ting
JPY
,
Kenney
SC
. 
Inhibition of IFN-gamma signaling by an Epstein-Barr virus immediate-early protein
.
Immunity
2001
;
15
:
787
99
.
22.
Zuo
JM
,
Thomas
WA
,
Haigh
TA
,
Fitzsimmons
L
,
Long
HM
,
Hislop
AD
, et al
Epstein-Barr virus evades CD4(+) T cell responses in lytic cycle through BZLF1-mediated downregulation of CD74 and the cooperation of vBcl-2
.
Plos Pathog
2001
;
7
:
e1002455
.
23.
Ma
SD
,
Hegde
S
,
Young
KH
,
Sullivan
R
,
Rajesh
D
,
Zhou
Y
, et al
A new model of Epstein-Barr virus infection reveals an important role for early lytic viral protein expression in the development of lymphomas
.
J Virol
2011
;
85
:
165
77
.
24.
Bogedain
C
,
Wolf
H
,
Modrow
S
,
Stuber
G
,
Jilg
W
. 
Specific cytotoxic T-lymphocytes recognize the immediate-early transactivator Zta of Epstein-Barr-Virus
.
J Virol
1995
;
69
:
4872
9
.
25.
Precopio
ML
,
Sullivan
JL
,
Willard
C
,
Somasundaran
M
,
Luzuriaga
K
. 
Differential kinetics and specificity of EBV-specific CD4(+) and CD8(+) T cells during primary infection
.
J Immunol
2003
;
170
:
2590
8
.
26.
Scherrenburg
J
,
Piriou
ER
,
Nanlohy
NM
,
van Baarle
D
. 
Detailed analysis of Epstein-Barr virus-specific CD4+ and CD8+ T cell responses during infectious mononucleosis
.
Clin Exp Immunol
2008
;
153
:
231
9
.
27.
Baiocchi
RA
,
Ward
JS
,
Carrodeguas
L
,
Eisenbeis
CF
,
Peng
R
,
Roychowdhury
S
, et al
GM-CSF and IL-2 induce specific cellular immunity and provide protection against Epstein-Barr virus lymphoproliferative disorder
.
J Clin Invest
2001
;
108
:
887
94
.
28.
Bosma
GC
,
Fried
M
,
Custer
RP
,
Carroll
A
,
Gibson
DM
,
Bosma
MJ
. 
Evidence of functional lymphocytes in some (Leaky) SCID mice
.
J Exp Med
1988
;
167
:
1016
33
.
29.
Mosier
DE
,
Gulizia
RJ
,
Baird
SM
,
Wilson
DB
. 
Transfer of a functional human immune-system to mice with severe combined immunodeficiency
.
Nature
1988
;
335
:
256
9
.
30.
Rowe
M
,
Young
LS
,
Crocker
J
,
Stokes
H
,
Henderson
S
,
Rickinson
AB
. 
Epstein-Barr-Virus (EBV)-associated lymphoproliferative disease in the SCID mouse model - implications for the pathogenesis of EBV-positive lymphomas in man
.
J Exp Med
1991
;
173
:
147
58
.
31.
Clark
KR
,
Voulgaropoulou
F
,
Fraley
DM
,
Johnson
PR
. 
Cell-lines for the production of recombinant adenoassociated virus
.
Hum Gene Ther
1995
;
6
:
1329
41
.
32.
Clark
KR
,
Sferra
TJ
,
Johnson
PR
. 
Recombinant adeno-associated viral vectors mediate long-term transgene expression in muscle
.
Hum Gene Ther
1997
;
8
:
659
69
.
33.
Clark
KR
,
Liu
XL
,
McGrath
JP
,
Johnson
PR
. 
Highly purified recombinant adeno-associated virus vectors are biologically active and free of detectable helper and wild-type viruses
.
Hum Gene Ther
1999
;
10
:
1031
9
.
34.
Clark
KR
. 
Recent advances in recombinant adeno-associated virus vector production
.
Kidney Int
2002
;
61
:
S9
15
.
35.
Alinari
L
,
Yu
B
,
Christian
BA
,
Yan
F
,
Shin
J
,
Lapalombella
R
, et al
Combination anti-CD74 (milatuzumab) and anti-CD20 (rituximab) monoclonal antibody therapy has in vitro and in vivo activity in mantle cell lymphoma
.
Blood
2011
;
117
:
4530
41
.
36.
Paquette
RL
,
Hsu
NC
,
Kiertscher
SM
,
Park
AN
,
Tran
L
,
Roth
MD
, et al
Interferon-alpha and granulocyte-macrophage colony-stimulating factor differentiate peripheral blood monocytes into potent antigen-presenting cells
.
J Leukoc Biol
1998
;
64
:
358
67
.
37.
Baiocchi
RA
,
Caligiuri
MA
. 
Low-dose interleukin 2 prevents the development of Epstein-Barr virus (EBV)-associated lymphoproliferative disease in SCID/SCID mice reconstituted i.p. with EBV-seropositive human peripheral blood lymphocytes
.
Proc Natl Acad Sci U S A
1994
;
91
:
5577
81
.
38.
Fuzzati-Armentero
MT
,
Duchosal
MA
. 
hu-PBL-SCID mice: an in vivo model of Epstein-Barr virus-dependent lymphoproliferative disease
.
Histol Histopathol
1998
;
13
:
155
68
.
39.
Keating
S
,
Prince
S
,
Jones
M
,
Rowe
M
. 
The lytic cycle of Epstein-Barr virus is associated with decreased expression of cell surface major histocompatibility complex class I and class II molecules
.
J Virol
2002
;
76
:
8179
88
.
40.
Porcu
P
,
Eisenbeis
CF
,
Pelletier
RP
,
Davies
EA
,
Baiocchi
RA
,
Roychowdhury
S
, et al
Successful treatment of posttransplantation lymphoproliferative disorder (PTLD) following renal allografting is associated with sustained CD8(+) T-cell restoration
.
Blood
2002
;
100
:
2341
8
.
41.
Tary-Lehmann
M
,
Saxon
A
. 
Human mature T cells that are anergic in vivo prevail in SCID mice reconstituted with human peripheral blood
.
J Exp Med
1992
;
175
:
503
16
.
42.
Gottschalk
S
,
Ng
CY
,
Perez
M
,
Smith
CA
,
Sample
C
,
Brenner
MK
, et al
An Epstein-Barr virus deletion mutant associated with fatal lymphoproliferative disease unresponsive to therapy with virus-specific CTLs
.
Blood
2001
;
97
:
835
43
.
43.
Khanim
F
,
Yao
QY
,
Niedobitek
G
,
Sihota
S
,
Rickinson
AB
,
Young
LS
. 
Analysis of Epstein-Barr virus gene polymorphisms in normal donors and in virus-associated tumors from different geographic locations
.
Blood
1996
;
88
:
3491
501
.
44.
Farrell
PJ
. 
Role for HLA in susceptibility to infectious mononucleosis
.
J Clin Invest
2007
;
117
:
2756
8
.
45.
Disis
ML
,
Bernhard
H
,
Shiota
FM
,
Hand
SL
,
Gralow
JR
,
Huseby
ES
, et al
Granulocyte-macrophage colony-stimulating factor: an effective adjuvant for protein and peptide-based vaccines
.
Blood
1996
;
88
:
202
10
.
46.
Mach
N
,
Gillessen
S
,
Wilson
SB
,
Sheehan
C
,
Mihm
M
,
Dranoff
G
. 
Differences in dendritic cells stimulated in vivo by tumors engineered to secrete granulocyte-macrophage colony-stimulating factor or flt3-ligand
.
Cancer Res
2000
;
60
:
3239
46
.
47.
Serafini
P
,
Carbley
R
,
Noonan
KA
,
Tan
G
,
Bronte
V
,
Borrello
I
. 
High-dose granulocyte-macrophage colony-stimulating factor-producing vaccines impair the immune response through the recruitment of myeloid suppressor cells
.
Cancer Res
2004
;
64
:
6337
43
.
48.
Lustberg
ME
,
Pelletier
RP
,
Porcu
P
,
Martin
SI
,
Quinion
CD
,
Geyer
SM
, et al
Human leukocyte antigen type and posttransplant lymphoproliferative disorder
.
Transplantation
2015
;
99
:
1220
5
.