Cell-mediated immunity, especially the cytotoxic T lymphocyte (CTL), provides resistance to Epstein-Barr virus (EBV), as is demonstrated by the occurrence of posttransplant lymphoproliferative disease in immunosuppressed patients. We set out to use dendritic cells (DCs) to elicit anti–EBV-specific CTLs in culture. In unselected, HLA-B8+ donors, monocyte-derived mature DCs were pulsed with the HLA-B8–restricted EBNA-3A peptide, FLRGRAYGL, and added to autologous T cells for 7 days at a DC:T ratio of 1:5 to 1:60. The cultured cells specifically lysed EBNA-3A peptide-pulsed, HLA-B8+, B-lymphoblastoid cell lines in a 5-hour51Cr-release assay. The generation of CTLs did not require the addition of interleukin-2. In comparison, monocytes were weak antigen-presenting cells. DCs were then infected with recombinant vaccinia-EBNA-3A. Vaccinia infection significantly decreased the viability of immature DCs after 3 days of culture (to 25% to 45%) but had a smaller effect on mature DC recovery (40% to 70%). To decrease these cytopathic effects and to expand the potential use of vaccinia vectors for DC therapy in immunocompromised patients, we successfully used psoralen and UV-inactivated virus. Mature DCs pulsed with either live or inactivated vaccinia EBNA-3A virus could elicit strong EBNA-3A–specific CTLs. Therefore, mature DCs are powerful stimulators of EBV-specific CTLs and their major histocompatibility complex class I products can even be charged with UV-inactivated recombinant vaccinia.

EPSTEIN-BARR VIRUS (EBV) is a ubiquitous human γ-herpes virus that has been associated with several malignant diseases. These diseases include nasopharyngeal carcinoma, Burkitt’s lymphoma, approximately 50% of Hodgkin’s disease, and lymphoproliferative disorders in the immunocompromised patient.1,2 Posttransplant lymphoproliferative disorders (PTLPD) of B cells can develop in the setting of organ transplants and in hematopoietic stem cell transplantation; for the latter, the incidence can be as high as 10%.3 Complete regression of PTLPD has been reported in 40% of patients after reduction or discontinuation of immunosuppressive therapy, but this is less feasible in marrow transplantation, because a likely result is a flare of graft-versus-host disease. These findings, as well as the observation of clinically apparent virus replicative lesions in T-cell immunocompromised patients,4 strongly suggest an important role for cell-mediated immune responses in the control of EBV. Further evidence comes from studies in which enriched populations of EBV-specific cytotoxic T lymphocytes (CTLs), generated from normal donors, are adoptively transferred to bone marrow transplant patients.5-8 The transferred cells provide efficient prophylaxis and demonstrable treatment of immunoblastic lymphoma.

We wanted to assess the feasibility of using dendritic cells (DCs) to elicit CTL responses to EBV. DCs are the most potent antigen-presenting cells (APCs),9,10 and their role in resistance against experimental malignancies11-18 and infections19 20 is well described. It is now possible to generate large numbers of DCs from bone marrow, cord blood, and peripheral blood. If DCs could elicit EBV-specific CTL responses, this would be advantagous for generating CTL lines, because DCs can be generated in a much shorter time frame than the EBV-transformed lymphoblastoid cell lines (LCL) that are now in use. Also, DCs could potentially be used to actively boost a patient’s EBV-specific immunity, in contrast to passive transfer of chronically stimulated T-cell lines.

EBV establishes a growth-transforming infection of B lymphocytes. Infection is associated with the expression of 6 virus-encoded nuclear antigens (EBNA-1, -2, -3A, -3B, -3C, and -LP) and 2 latent membrane proteins (LMP-1 and -2). The primary and memory CD8+ CTL response in healthy EBV carriers is markedly skewed toward HLA allele-specific epitopes drawn from the EBNA-3A, -3B, and -3C subset of latent proteins.21-23 Reactivities to other EBV latent antigens are less frequent. In lymphoproliferative disorders in the immunocompromised patient, the full array of latent EBV antigens is expressed.24,25 In this study, we wanted to investigate the use of DCs to generate CTL responses in HLA-B8+, healthy, EBV+ carriers to the immunodominant EBV antigen EBNA-3A in a relatively short culture assay of 7 days. To initially test the use of DCs, we used the well-described HLA-B8+ T-cell epitope FLRGRAYGL from the EBNA-3A antigen. Then, to expand this method for use with other HLA-types and EBV antigens, we tested recombinant vaccinia virus as a source of antigen.21 26 We will show that DCs can be strong stimulators of EBV-specific CTL responses in culture and, remarkably, that UV-inactivated recombinant vaccinia virus can serve as a source of EBV antigen.

Culture Medium

The medium used for generation of DCs and for CTL induction was RPMI-1640 supplemented with 10 mmol/L HEPES, 5 mmol/L L-glutamine, 20 μg/mL of gentamicin, and either 1% plasma (heparinized) or 5% pooled or single donor human serum, heat-inactivated for 30 minutes at 56°C.

Cytokines

We purchased recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF; Sargramostim Leukine; 1.4 × 106 U/250 μg) from Immunex (Seattle, WA), recombinant interleukin-4 (IL-4; 4.1 × 107 U/mg) from Genzyme (Cambridge, MA), and IL-2 from Schiapparelli Biosystems (Fairfield, NJ). Lymphocult was puchased from Biotest (Dreieich, Germany).

Cell Lines

EBV-transformed B-LCLs were established by culturing peripheral blood mononuclear cells (PBMCs) of HLA class I-typed donors with supernatant from the marmoset line B95.8 in the presence of 1 μg/mL cyclosporin A in medium supplemented with 20% fetal calf serum (FCS). The TAP−/−, HLA-A2.1+ T2 cell line from the American Type Culture Collection (ATCC; Manassas, VA) was used as targets for testing the cytotoxic activity of influenza-specific CTLs. The BSC40 monkey kidney line (ATCC) was grown in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 5% FCS and used in plaque assays to titer recombinant vaccinia virus stocks. The RK13 rabbit kidney line (ATCC) was grown in DMEM supplemented with 15% FCS and used for expansion of the vaccinia virus stock.

Mononuclear Cell Subsets

PBMCs.

PBMCs were isolated from leukocyte-enriched buffy coats by standard density gradient centrifugation on Ficoll-Paque (Pharmacia, Uppsala, Sweden). T-cell–enriched (ER+) and T-cell–depleted (ER) populations were prepared by rosetting with neuraminidase-treated (Vibrio cholerae neuraminidase; Calbiochem, La Jolla, CA) sheep red blood cells (Colorado Serum Co, Denver, CO).

T cells.

E-rosetted T cells were further purified by removal of monocytes, natural killer (NK) cells, and major histocompatibility complex (MHC) class II-positive cells by panning with antibodies to CD11b, CD16, and HLA-DR, as described.27 

DCs.

A total of 2.5 × 106 ER cells were plated in 3 mL volumes in 6-well tissue culture dishes (Falcon, Lincoln Park, NJ) in complete medium containing 1% human plasma. GM-CSF and IL-4 were added at final concentrations of 500 U/mL of IL-4 and 1,000 U/mL of GM-CSF. Cytokines and medium were replenished on days 2, 4, and 6.28,29 On day 7, nonadherent cells were collected and transferred to new 6-well plates. The cultures were supplemented with monocyte conditioned medium (MCM; final concentration, 50% vol/vol) to induce final maturation of the DCs that were harvested 48 hours later.30 

Vaccinia Virus

Virus stocks.

We used recombinant vaccinia virus (rVV), expressing the EBV latent gene EBNA-3A or LMP-121 or the influenza matrix gene, vJL3 (vJL3 was kindly provided to us by B. Moss, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD).31 The control was the parental vaccinia virus construct that is negative for thymidine kinase (rVV-TK).26 

Vaccinia virus expansion.

Vaccinia virus stocks were expanded using adherent rabbit kidney cells, RK13 cells. Contaminating vaccinia proteins were digested with trypsin (GIBCO BRL, Life Technologies, Grand Island, NY). The virus preparation also was centrifuged through a sucrose gradient to further remove proteins and peptides.

Virus inactivation.

A psoralen (Sigma, Steinheim, Germany) stock solution (1 mg/mL, 50% H2O/50% ethanol) was added to the viruses (2 × 106 to 1 × 107 plaque-forming units [PFU]) at a concentration of 10 μg/mL in a flat-bottom 96-well plate (Costar, Cambridge, MA) and incubated for 10 minutes at room temperature. Then, to inactivate the virus, the preparation was irradiated in a Stratalinker 1800 UV cross-linking unit (Stratagene, La Jolla, CA) equipped with five 365-nm UV bulbs for 6 minutes (PLWUV, psoralen long wave UV).32 

Viral titers.

Plaque-forming activities of active and inactivated virus were determined by serial dilutions on a BSC40 cell layer. Plaques were counted after rVV infection of BSC40 cells at a titration starting from 109 PFU/mL to 103 PFU/mL, in duplicates in 6-well plates. A titer of 0 was recorded when no plaques formed on the BSC40 cell monolayer after 2 days.

Influenza Virus

Influenza A virus (PR8, Puerto Rico/8/34; source: allantoic fluid) was purchased from Spafas Inc (Storrs, CT).

Antigens

Synthetic peptides.

The EBNA-3A peptides, FLRGRAYGL and QAKWRLQTL, were purchased from Biosynthesis (Lewisville, TX). The EBNA-3A peptide FLRGRAYGI was purchased from Genemed Synthesis (San Francisco, CA). All peptides were greater than 95% pure by mass spectrometry and high-performance liquid chromatography (HPLC). Stock solutions in dimethyl sulfoxide (DMSO) were kept at −70°C. The influenza A virus matrix peptide GILGFVFTL was used to pulse T2 cells for 51Cr release assays.

Antigen-pulsing of DCs.

DCs were harvested out of the 6-well plates after 24 or 48 hours of MCM and resuspended in 1% plasma at 1 × 107/mL. rVV was added at a multiplicity of infection (MOI) of 1:1 or 2:1 and was incubated for 1 hour at 37°C. DCs were infected with influenza virus in serum-free RPMI for 1 hour at 37°C at an MOI of 0.5:1. Peptide pulsing of mature DCs was performed for 1 hour in RPMI at room temperature at a final concentration of 10 μmol/L. DCs were washed 3 times and used to stimulate bulk cultures of purified syngeneic T cells in 96- or 24-well plates (Costar) at DC to T-cell ratios of 1:5 to 1:60.

Fluorescence-Activated Cell Sorting (FACS) Analysis of Cell Populations and Vaccinia Infection

Serological HLA class I typing.

Buffy coats were typed with monoclonal antibodies (MoAbs) to HLA-B8 (One Lambda, Canoga Park, CA) and HLA-A2 (ATCC) and analyzed by FACScan. Target LCL lines were HLA-typed at Memorial Sloan Kettering Cancer Center (New York, NY).

T cells.

T cells were phenotyped by staining with Simultest CD4-fluorescein isothiocyanate (FITC)/CD8-phycoerythrin (PE) or Simultest control IgG1-FITC/IgG2a-PE from Becton Dickinson (BD; San Jose, CA). To document purity of the panned T cells, we verified the absence of cells that stained for CD56-PE (BD) and HLA-DR-PE (BD), prepanning and postpanning.

DCs.

MoAbs to the following surface antigens were used: HLA-DR-PE, CD14-PE, CD25-PE (all BD), CD86-PE (PharMingen, San Diego, CA), CD83-PE (Coulter Corp, Miami, FL), and the antivaccinia hemagglutinin antibody VV1-4G9. PE-conjugated F(ab′)2 goat antimouse IgG (γ and light chain; Tago, Burlingame, CA) was used as a secondary antibody. Cells were phenotyped with the panel of MoAbs and analyzed on a FACScan.

Annexin V/propidium iodide (PI) staining.

Annexin V/PI staining (Kamiya, Seattle, WA) was used to monitor cytopathic effects on uninfected DCs and DCs infected with live and PLWUV-inactivated rVV. The DCs were plated at 1 × 105/200 μL of 5% pooled human serum per well in flat-bottom 96-well plates. Each day after infection, the cells were stained with 5 μL of Annexin V and 5 μL of PI and immediately analyzed on a FACScan.

Intracellular staining.

DCs and B-LCLs were fixed with 4% paraformaldehyde, washed, and permeabilized with 1% saponin for 30 minutes at 4°C. Anti-LMP-133 (Dako, Glostrup, Denmark) and antivaccinia antibodies (VV1-2F10, VV3-5B8, VV4-2F6, and VV1-6B6) were added for 30 minutes. Cells were washed in phosphate-buffered saline (PBS) containing 0.1% saponin, 0.1% azide, 1% FCS, and 1% human serum and then incubated with 1:250 PE- or 1:100 FITC-conjugated goat antimouse IgG (Biosource, Camarillo, CA). Cells were washed twice and analyzed by FACScan.

T-Cell Responses

Allogeneic MLR.

Uninfected and infected DCs were added in graded doses as stimulators for 2 × 105 purified, allogeneic T cells in 96-well flat-bottom plates. Proliferation was determined on day 5 with the addition of 4 μCi/mL of 3H-TdR for 8 to 12 hours to triplicate wells.

Induction of CTL responses.

A total of 2 × 105 or 1 × 106purified T cells were cultured in 96- or 24-well plates with graded doses of peptide-pulsed DCs or rVV-infected or uninfected DCs, in a total volume of 200 μL or 1 mL of 5% single donor serum. In 4 of 6 experiments using FLRGRAYGL-pulsed DCs as stimulators, cultures were set up with and without the addition of IL-2 (50 IU/mL) on day 3 postsetup. When using the QAKWRLQTL peptide, longer culture times were required to detect strong CTLs. T cells were restimulated with peptide-pulsed DCs on day 7, Lymphocult was added on days 1 and 7, and the cultures were harvested on day 14. To assay CTLs, cells from the 24-well plates were transferred to 96-well plates.

51Cr release assay for effector CTLs.

LCLs and T2 cells were incubated with peptide (10 μmol/L) for 1 hour at room temperature and then labeled for 1 hour with 400 μCi of Na51CrO4 (1 mCi/mL, sterile stock; New England Nuclear, Boston, MA) at 37°C. The cells were washed 4 times and resuspended at 2 × 105/mL, and 1 × 104 target cells were added to each well of a 96-well plate to give effector:target ratios of 10:1 to 30:1. Spontaneous and total release samples were prepared by adding the targets to wells containing RPMI alone or a final concentration of 0.33% sodium dodecyl sulfate (SDS), respectively. The plates were centrifuged for 2 minutes at 15g and incubated for 5 hours at 37°C. At the termination of the assay, the supernatants were collected with absorption cartridges using a harvesting press (Skatron Instruments Inc, Sterling, VA) and counted in a γ-counter. All tests were conducted in triplicate, and the percentage of specific lysis was calculated from the following formula: 100 × ([release by CTL − spontaneous release]/[total release − spontaneous release]). Spontaneous release was 15% to 25% of the total release.

CTL Induction With EBNA-3A (FLRGRAYGL) Peptide-Pulsed DCs

Peptide-pulsed and unpulsed DCs were added to autologous T cells and, after 7 days, the cultured cells were tested for killing activity on HLA-matched (B8+) and mismatched (A2+) LCL lines in a standard 5-hour 51Chromium release assay. In 6 of 6 HLA-B8+ donors, mature DCs that were pulsed with the HLA-B8+ dominant peptide FLRGRAYGL induced strong CTL responses in culture (Table 1). Effector:target ratios of 10:1 to 20:1 were sufficient to detect specific lysis of HLA-matched LCLs, and the killing was restricted to B8+ targets (Fig 1A). Different ratios of DCs to T cells were tested. A DC:T-cell ratio of 1:60 was sufficient to induce CTLs (Table 1). In 4 of 6 experiments, IL-2 (50 IU/mL) was added on day 3 of cultures, but this was not required for DCs to elicit CTL responses. High CTL responses were not further increased through the addition of IL-2, but weaker responses could be enhanced (Table 1 and Fig 1B). As has been described before,34,35 B-LCL could only serve as a CTL target if pulsed with exogenous peptide. The B-LCLs used throughout our experiments were transformed with the viral strain B95.8, which carries sequence variations in the EBNA-3A gene compared with the EBV strain A found in the western hemisphere, eg, the Leucine in the FLRGRAYGL peptide is mutated to Isoleucine in B95.8.36 Therefore, we used another peptide (QAKWRLQTL) that is the same in the different viral strains to stimulate syngeneic T cells (Fig 1C). In repeated experiments using different peptide concentrations for pulsing our DCs (10 μmol/L to 100 nmol/L, data not shown), we still could not detect lysis of the B-LCLs alone. The B-LCLs had to be pulsed with exogenous peptide to serve as targets.

Table 1.

A Summary of the First 6 HLA-B8+ Persons in Which DCs Were Pulsed With EBNA-3A Peptide, Added at a Dose of 1:5 to 1:60 to T Cells, Without or With Supplemental IL-2

Experiment No. E:T RatioDC:T Ratio % Specific Lysis
Without IL-2 With IL-2
B8+ LCL, No Peptide B8+LCL+ FLRGRAYGL Peptide B8+ LCL, No Peptide B8+ LCL+ FLRGRAYGL Peptide
1  10:1  1:5   0  41  NT  NT 
 20:1  1:5   0  75  NT  NT  
2  10:1 1:10  0  90   0  96  
  1:30  0  76  5  96  
3  20:1  1:38  7  31  NT  NT  
20:1  1:30  0  12   5  22  
5  20:1  1:60 0  11   5  23  
6  10:1  1:60  NT  NT  4  60  
 20:1  1:60  5  68  19  71 
Experiment No. E:T RatioDC:T Ratio % Specific Lysis
Without IL-2 With IL-2
B8+ LCL, No Peptide B8+LCL+ FLRGRAYGL Peptide B8+ LCL, No Peptide B8+ LCL+ FLRGRAYGL Peptide
1  10:1  1:5   0  41  NT  NT 
 20:1  1:5   0  75  NT  NT  
2  10:1 1:10  0  90   0  96  
  1:30  0  76  5  96  
3  20:1  1:38  7  31  NT  NT  
20:1  1:30  0  12   5  22  
5  20:1  1:60 0  11   5  23  
6  10:1  1:60  NT  NT  4  60  
 20:1  1:60  5  68  19  71 

Seven days later, CTLs were measured on B8+ LCLs to which EBNA-3A peptide was or was not added. The left-hand column is the T cell (E):target cell (T) ratio for CTL assay.

Abbreviation: NT, not tested.

Fig. 1.

Induction of EBV-specific CTLs with peptide-pulsed DCs. (A) Peptide (FLRGRAYGL)-pulsed DCs induce HLA class I-restricted lysis of peptide-pulsed B8+ LCLs after 7 days of coculture. The CTLs did not lyse unpulsed HLA-matched LCL or peptide-pulsed B8 LCL. The effector:target ratio was 20:1. (B) When the induction of CTL was relatively low, the response could be enhanced through the addition of IL-2 (with 50 IU/mL) on day 3 of the 7-day culture. (C) QAKWRLQTL-pulsed DCs induce HLA class I-restricted lysis of peptide-pulsed B8+ LCLs after 14 days of coculture. The culture was restimulated with peptide pulsed DCs on day 7 and IL-2 was added on days 1 and 7. Two effector:target ratios are shown (10:1 and 30:1).

Fig. 1.

Induction of EBV-specific CTLs with peptide-pulsed DCs. (A) Peptide (FLRGRAYGL)-pulsed DCs induce HLA class I-restricted lysis of peptide-pulsed B8+ LCLs after 7 days of coculture. The CTLs did not lyse unpulsed HLA-matched LCL or peptide-pulsed B8 LCL. The effector:target ratio was 20:1. (B) When the induction of CTL was relatively low, the response could be enhanced through the addition of IL-2 (with 50 IU/mL) on day 3 of the 7-day culture. (C) QAKWRLQTL-pulsed DCs induce HLA class I-restricted lysis of peptide-pulsed B8+ LCLs after 14 days of coculture. The culture was restimulated with peptide pulsed DCs on day 7 and IL-2 was added on days 1 and 7. Two effector:target ratios are shown (10:1 and 30:1).

Close modal

Live and UV-Inactivated Vaccinia Infection of DCs

Immature (no addition of MCM) and mature DCs were infected with rVV at an MOI of 2:1. Expression of viral protein was monitored by FACS with a panel of MoAbs reacting with 1 early and 2 late proteins in the vaccinia replication cycle. These IgG2a antibodies stain an early vaccinia virus protein (VV1-6B6), the late vaccinia hemagglutinin (VV1-4G9), and the late D8L virion/surface protein (VV4-4G9; Table 2). When all of the antibodies were tested on rVV infected B-LCL and DCs, the B-LCLs expressed early and late vaccinia antigens, whereas DCs only expressed early antigens (Fig 2A). We also stained rVV-LMP-1–infected DCs with LMP-1 antibody and the VV1-6B6 antibody. Both proteins were expressed under the control of the early promoters and resulted in the same staining pattern (Fig 2B). The VV1-6B6 early antigen was used in all of our experiments to monitor the percentage of infected DCs.

Table 2.

Mouse Antivaccinia MoAbs Used for FACS Analysis of Recombinant Vaccinia Virus Infection of DCs and B-LCLs

Clone Isotype Reaction With
VV1-4G9  IgG2a Vaccinia hemagglutinin (cell surface, late)  
VV4-2F6 IgG2a  Reacts with D8L, a virion-surface protein 
VV1-6B6  IgG2a  Reacts with an early protein of ∼29 kD 
Clone Isotype Reaction With
VV1-4G9  IgG2a Vaccinia hemagglutinin (cell surface, late)  
VV4-2F6 IgG2a  Reacts with D8L, a virion-surface protein 
VV1-6B6  IgG2a  Reacts with an early protein of ∼29 kD 

MoAbs from mouse ascites, at a concentration of 2 mg/mL or greater, were used at a 1:10,000 dilution, which was optimal.

Fig. 2.

Antibody staining 16 hours after infection of mature DCs with rVV. (A) Infection of DCs and LCL was compared using FACS staining (y-axis) and MoAbs against early and late vaccinia virus proteins (Table 2). VV1-4G9 was a surface stain, whereas all other antibodies required cell permeabilization. IgG2a was used as an isotype control. Infected DCs (arrows) only stained for the early vaccinia virus protein recognized by VV1-6B6, whereas infected LCL (arrows) stained for early and late vaccinia virus proteins. (B) The VV1-6B6 antibody was compared with the LMP-1 antibody, after infection of mature DCs with rVV-LMP-1. The VV1-6B6 and LMP-1 are early proteins, with the latter being an EBV protein. Both antibodies stained the same percentage of DCs, thereby confirming the accuracy of the VV1-6B6 antibody.

Fig. 2.

Antibody staining 16 hours after infection of mature DCs with rVV. (A) Infection of DCs and LCL was compared using FACS staining (y-axis) and MoAbs against early and late vaccinia virus proteins (Table 2). VV1-4G9 was a surface stain, whereas all other antibodies required cell permeabilization. IgG2a was used as an isotype control. Infected DCs (arrows) only stained for the early vaccinia virus protein recognized by VV1-6B6, whereas infected LCL (arrows) stained for early and late vaccinia virus proteins. (B) The VV1-6B6 antibody was compared with the LMP-1 antibody, after infection of mature DCs with rVV-LMP-1. The VV1-6B6 and LMP-1 are early proteins, with the latter being an EBV protein. Both antibodies stained the same percentage of DCs, thereby confirming the accuracy of the VV1-6B6 antibody.

Close modal

Immature DCs were infected at a higher frequency (40% to 80%) than mature DCs (15% to 65%). DC viability was tested 1 to 5 days postinfection with trypan blue exclusion (Fig 3A) and Annexin V/PI FACS staining (Fig 3B). Viability was decreased relative to uninfected DCs. For immature DCs, viable cell recovery was 25% to 45%, whereas for mature DCs, recovery was 40% to 70% on day 3 after infection (Fig 3A). Vaccinia virus is a known cytopathic virus, and, as the results from Annexin V/PI staining indicated, infection led to an increase in the number of DCs undergoing apoptosis (Fig 3B). In parallel studies, we have noted that, at early time points, vaccinia virus infection blocks the maturation of immature DCs.37 

Fig. 3.

Cytotoxicity of rVV towards mature DCs. (A) DCs were recultured at 1 × 105/200 μL of 5% human serum in 1 well of a 96-well flat-bottom well plate, and cell viability was tested with trypan blue staining for 5 days. Live and PLWUV-inactivated, rVV-infected, and uninfected DCs were compared. Cell viability of rVV-infected mature DCs decreased significantly by day 3 compared with PLWUV rVV-infected and uninfected DCs. (B) Viability of uninfected and infected mature DCs was monitored with Annexin V and PI staining (PI stain not shown). The VV1-6B6 antibody indicated the rate of infection. Mature DCs were infected with live and PLWUV-inactivated rVV-LMP-1 at an MOI of 2:1. Twenty-four to 30 hours later, Annexin V FITC staining was performed and immediately analyzed by FACScan. DCs infected with live virus showed greater than 30% Annexin V+ cells, whereas uninfected and PLWUV rVV-infected DCs showed less than 10% positive cells.

Fig. 3.

Cytotoxicity of rVV towards mature DCs. (A) DCs were recultured at 1 × 105/200 μL of 5% human serum in 1 well of a 96-well flat-bottom well plate, and cell viability was tested with trypan blue staining for 5 days. Live and PLWUV-inactivated, rVV-infected, and uninfected DCs were compared. Cell viability of rVV-infected mature DCs decreased significantly by day 3 compared with PLWUV rVV-infected and uninfected DCs. (B) Viability of uninfected and infected mature DCs was monitored with Annexin V and PI staining (PI stain not shown). The VV1-6B6 antibody indicated the rate of infection. Mature DCs were infected with live and PLWUV-inactivated rVV-LMP-1 at an MOI of 2:1. Twenty-four to 30 hours later, Annexin V FITC staining was performed and immediately analyzed by FACScan. DCs infected with live virus showed greater than 30% Annexin V+ cells, whereas uninfected and PLWUV rVV-infected DCs showed less than 10% positive cells.

Close modal

To decrease the cytopathic effect of vaccinia virus on DCs and to expand the potential use of vaccinia vectors in immunocompromised patients, we used psoralen and UVA-inactivated, nonreplicating vaccinia virus to infect mature DCs. Inactivation of rVV was monitored by a plaque-forming assay with BSC40 cells, as well as with FACS staining to follow synthesis of vaccinia virus proteins. Plaques were counted after infection with live and UV-inactivated rVV at a titration starting at 109 PFU/mL to 103 PFU/mL, in duplicates in 6-well plates. There were no plaques detectable with PLWUV-inactivated rVV in 3 separate experiments. To prevent protein or peptide contamination, viral preparations were digested with trypsin and centrifuged through a sucrose gradient. After infection of DCs with PLWUV-inactivated virus, an MoAb against an early vaccinia protein (VV1-6B6) gave a decreased staining (0% to 15%) compared with live rVV infected DCs (Fig 3B).

Mature DCs that were infected with live or PLWUV-inactivated rVV had the same phenotype and function as uninfected DCs. Specifically, infected and uninfected DCs expressed high levels of CD83, CD86, HLA-DR, and CD25, as is typical of mature DCs (Fig 4A), and these DCs were potent stimulators of allogeneic T-cell responses (Fig 4B). The efficacy of rVV-infected DCs in stimulating the MLR might reflect DC function before their death from infection or the capacity of the responding T cell to rescue the DC.

Fig. 4.

FACS analysis of rVV-infected mature DCs. (A) The top row shows staining for VV1-6B6 early vaccinia protein in uninfected, PLWUV rVV-TK and live rVV-TK-infected DCs at an MOI of 2:1. The lower row shows comparable staining for CD25 and CD83, 2 markers for DC maturation, on uninfected DCs (···), PLWUV rVV-infected DCs (—-), and rVV-infected DCs (—). (B) rVV-infected DCs stimulate T-cell proliferative responses in the MLR. Bulk T cells were used as responders towards graded doses of allogeneic DCs that were uninfected or infected with rVV-TK and PLWUV rVV-TK. Cultures were pulsed on day 5 for 8 hours with 4 μCi/mL of 3H-TdR. Results are the means of triplicates (mean cpm).

Fig. 4.

FACS analysis of rVV-infected mature DCs. (A) The top row shows staining for VV1-6B6 early vaccinia protein in uninfected, PLWUV rVV-TK and live rVV-TK-infected DCs at an MOI of 2:1. The lower row shows comparable staining for CD25 and CD83, 2 markers for DC maturation, on uninfected DCs (···), PLWUV rVV-infected DCs (—-), and rVV-infected DCs (—). (B) rVV-infected DCs stimulate T-cell proliferative responses in the MLR. Bulk T cells were used as responders towards graded doses of allogeneic DCs that were uninfected or infected with rVV-TK and PLWUV rVV-TK. Cultures were pulsed on day 5 for 8 hours with 4 μCi/mL of 3H-TdR. Results are the means of triplicates (mean cpm).

Close modal

CTL Induction With DCs Infected With Live and PLWUV-Inactivated rVV

Because HLA-B8+ donors gave a reliable CTL response after stimulation with EBNA-3A peptide-pulsed DCs, we used live rVV-EBNA-3A to deliver antigens to the MHC class I products of mature DCs. In 3 HLA-B8+ positive random donors, we could elicit EBNA-3A–specific responses with vaccinia-infected DCs (Fig 5A). Because rVV-EBNA-3A is derived from the B95.8 viral strain, containing a mutated FLRGRAYGL peptide, we tested if rVV infected DCs could elicit CTLs against B-LCLs pulsed with either FLRGRAYGL or FLRGRAYGI (Fig 5B). rVV-EBNA-3A–infected DCs elicited good CTL responses in 7-day culture against both targets. Because FLRGRAYGI binds more weakly to the MHC class I molecule, the lysis was slightly decreased relative to FLRGRAYGL-pulsed B-LCLs.

Fig. 5.

CTL induction with peptide-pulsed and live rVV-infected DCs. (A) Comparison of EBNA-3A peptide-pulsed DCs with rVV-infected DCs. Live rVV-TK was used as negative control. (B) Comparion of monocytes and DCs as APCs in eliciting EBNA-3A–specific CTL responses in a 7-day culture assay. The top panel shows that rVV-EBNA-3A–infected DCs (DC:T cell ratio 1:30) stimulate T cells that recognize FLRGRAYGL- and FLRGRAYGI-pulsed HLA B8+ B-LCLs. The lower panel from the same experiment shows that rVV-EBNA-3A–infected monocytes (ER:T-cell ratio 1:2) are unable to stimulate T cells that recognize FLRGRAYGL- or FLRGRAYGI-pulsed HLA B8+ B-LCLs. (C) Comparison of EBNA-3A peptide-pulsed DCs with PLWUV, rVV-EBNA-3A–infected DCs; PLWUV rVV-TK was used as a negative control. (D) Comparison of live versus PLWUV rVV-EBNA-3A–infected DCs. (E) Comparison of DCs infected with live influenza A or with live or PLWUV rVV-influenza-matrix in eliciting responses to the immunodominant matrix peptide for HLA-A2.1. In all of the experiments, the DC:T-cell ratio was 1:30, the effector T-cell:target cell ratio was 1:20, and no IL-2 was used.

Fig. 5.

CTL induction with peptide-pulsed and live rVV-infected DCs. (A) Comparison of EBNA-3A peptide-pulsed DCs with rVV-infected DCs. Live rVV-TK was used as negative control. (B) Comparion of monocytes and DCs as APCs in eliciting EBNA-3A–specific CTL responses in a 7-day culture assay. The top panel shows that rVV-EBNA-3A–infected DCs (DC:T cell ratio 1:30) stimulate T cells that recognize FLRGRAYGL- and FLRGRAYGI-pulsed HLA B8+ B-LCLs. The lower panel from the same experiment shows that rVV-EBNA-3A–infected monocytes (ER:T-cell ratio 1:2) are unable to stimulate T cells that recognize FLRGRAYGL- or FLRGRAYGI-pulsed HLA B8+ B-LCLs. (C) Comparison of EBNA-3A peptide-pulsed DCs with PLWUV, rVV-EBNA-3A–infected DCs; PLWUV rVV-TK was used as a negative control. (D) Comparison of live versus PLWUV rVV-EBNA-3A–infected DCs. (E) Comparison of DCs infected with live influenza A or with live or PLWUV rVV-influenza-matrix in eliciting responses to the immunodominant matrix peptide for HLA-A2.1. In all of the experiments, the DC:T-cell ratio was 1:30, the effector T-cell:target cell ratio was 1:20, and no IL-2 was used.

Close modal

To evaluate the ability of other APCs to induce CTL responses, we used the ER fraction of the sheep-rosetted PBMCs, which consisted mainly of monocytes. In contrast to DCs, monocytes were not able to elicit peptide specific CTLs in a 7-day culture system (Fig5B).

We then tested DCs infected with PLWUV-inactivated rVV. These DCs were as effective as DCs infected with live vaccinia in stimulating EBNA-3A–specific CTLs (Fig 5B and C). To confirm the use of PLWUV rVV as a vector for delivering antigen to DCs, we compared live and PLWUV-inactivated rVV carrying the influenza matrix gene. As shown in Fig 5D, DCs infected with live and inactivated rVV induced a comparable matrix peptide-specific CTL response to influenza-infected DCs.

Several laboratories have described a strong CD8+ T-cell response in healthy EBV+ carriers to the latent antigens of EBV. The dominant response is directed to EBNA-3A, -3B, and -3C gene products.1,2,22,26 These conclusions are based on CTL responses that are generated by repetitive stimulation of bulk mononuclear cells with B-LCLs and IL-2, followed by limiting dilution clonal analysis. However, relatively little information is available on the APCs that are responsible for eliciting this response. The EBV-infected B cell is likely to be important, but we also feel that DCs need to be studied for at least 2 reasons. First, even though DCs are not known to express the CD21 CR2 complement receptor responsible for EBV binding to host cells, it is possible that DCs capture EBV antigens from B cells that have undergone apoptosis.38Second, delivery of EBV antigens to DCs, using viral vectors as an example, could provide a new route whereby DCs would be used in therapy to induce stronger immunity in patients suffering from EBV-associated malignancies, eg, PTLPD, Hodgkin’s disease, and nasopharyngeal carcinoma.

In this report, we have begun to analyze the capacity of DCs to elicit CD8+ CTL immunity. We have found that mature DCs, after exposure to EBNA-3A peptide or infection with rVV-EBNA-3A, serve as efficient stimulators of CTL responses from healthy blood donors. A comparison of monocytes and DCs in eliciting EBV-specific CTL responses showed that DCs are more potent in generating memory responses to a dominant EBNA-3A–restricted peptide. At this time, we know that B-LCL can serve as APCs, but they are less efficient than DCs on a per cell basis and also require addition of exogenous IL-2 (data not shown). In our experience, DCs elicit CTL responses without the addition of exogenous cytokines, presumably because the mature type of DC that we used is known to be capable of eliciting strong IL-2 and cytokine production directly from CD4+ T-cell–depleted CD8+ cells.39 40 

There remains a puzzling limitation to the CTLs that were generated with DCs, which is the same limitation that has also been described for T-cell clones generated with LCLs as APCs.34,35 The limitation is that EBNA-3A–specific CTLs do not lyse HLA-B8+ targets directly. We considered that our HLA-B8+ targets might express a different epitope than our FLRGRAYGL-pulsed DCs, because of known sequence variations between the donor’s EBV sequence and the laboratory B95.8 strain used to generate the B-LCL.36,41 However, the same findings were made in repeated experiments using the QAKWRLQTL peptide, which is conserved between the different viral strains. Again, the HLA-matched B-LCL targets had to be enhanced through peptide pulsing. Therefore, our results are similar to those of other reports34 35 that B-LCLs do not seem to present endogenous EBNA peptides in sufficient amounts.

There are several reports of the successful use of T-cell lines for the prophylaxis and treatment of PTLPD,6,8 as well as 1 report on the use of CTL lines in the treatment of patients with EBV+ Hodgkin’s disease.42 The successful reduction in lymphoma and EBV viral burden is likely due to the action of EBV-specific CTLs after this passive immunotherapy. In these studies, B-LCLs were used as APCs to stimulate the formation of CTLs, and this required 4 to 6 weeks. Instead, our data encourage the use of DCs for generating EBV-specific responses for passive immunotherapy. Monocyte-derived DCs can be reliably generated in 7 to 9 days, and these mature DCs induce strong, HLA-restricted CTLs in 7 days.

The other approach would be to use DCs for active rather than passive immunotherapy, ie, DCs would be charged with EBV antigens ex vivo and reinfused to directly immunize patients. Active immunization might provide longer lasting and stronger T-cell memory. The longevity of passively transferred immune cells varies in different reports. Walter et al43 followed cytomegalovirus (CMV)-specific T cells by analysis of rearranged T-cell receptor genes and detected the transferred cells for at least 12 weeks. Heslop et al7 showed persistence of EBV-specific T cells for 18 months after adoptive transfer and after restimulation of CTLs ex vivo. If DCs were used to actively immunize patients, the patient might develop long-term memory and be able to repeatedly generate CTLs after exposure to EBV antigens. In contrast, memory may not readily be established if one transfers chronically stimulated, effector type cells that are programmed to die quickly after withdrawal of cytokines or antigen encounter (antigen-induced cell death).

The use of EBV peptides to charge DCs with antigen has some severe limitations in the context of immunotherapy. First, the peptides need to be defined and tailored to the patient’s HLA haplotype. Second, a single peptide may elicit a form of immunity that is easy for tumors to escape, because most immunogenic peptides are not known to be sequences that are essential for the transforming ability of the virus. We considered here vaccinia vectors, because an excellent panel of recombinant vaccinia viruses exists, each encoding one of the 8 latent EBV genes.26 There are reports in which rVV has been used to deliver antigens to mouse DCs and human DCs.44,45Although vaccinia virus is a known cytopathic virus, none of the prior reports address the degree of toxicity that is encountered when using this virus. The toxicity of vaccinia virus is more prominent in immature DCs that are also infected to a greater extent than our current preparation of mature DCs. In addition, infection with rVV blocks the maturation of DCs before any detectable cytolysis.37 

We have infected B-LCLs and immature and mature DCs with rVV and monitored the rate of infection with vaccinia antibodies that react with early and late vaccinia proteins. The VV1-6B6 antibody stains a protein that is inferred to be under the control of an early promotor that becomes active immediately after infection, because necessary transcription factors are brought into the cells within the infectious particles. Infected B-LCLs and DCs stain positively for VV1-6B6, as well as anti–LMP-1 when using rVV-LMP-1, which is under the control of an early promotor. In agreement with others,46,47 we find that macrophages and DCs ex vivo are nonpermissive for vaccinia virus because no late viral proteins are produced. This could be confirmed by staining with the VV1-4G9 antibody, which stains the vaccinia hemagglutinin protein A56R (controlled by a late promoter) on infected B-LCLs but not on DCs.48 The same findings are evident with VV4-2F6 that stains the late vaccinia protein D8L.49Consequently, we have used VV1-6B6 to monitor the percentage of infected DCs. We observe consistently that immature DCs are infected to a higher degree than mature DCs. Testing cell viability with trypan blue exclusion up to 5 days after infection shows a significant decrease in the viability of immaturely infected DCs. To exclude the possibility that the MCM we have used to induce maturation is responsible for this toxicity, we have performed comparative studies with prostaglandin E2 (PGE2; 10 μg/mL) and tumor necrosis factor-α (1,000 IU/mL) to induce maturation. No difference in cell viability as well as percentage of infection of mature DCs has been observed (data not shown). Staining with Annexin V FITC and PI shows that rVV-infected DCs undergo apoptosis faster and to a higher percentage compared with uninfected DCs (Fig 3B). However, before cell death, the uninfected and infected mature DCs have the same phenotype and function, as detected by the capacity to stimulate allogeneic T cells (Fig 4). The mechanism underlying the nonpermissive state for virus replication in macrophages as well as DCs is not known. There is evidence that the initiator of the apoptosis signal is ds RNA.50 Clearly, the maturation state of the DC somehow influences the degree of infection with rVV.

To increase the possibility of using rVV in immunocompromised patients and to possibly decrease the cytopathic effect of vaccinia, we have used vaccinia inactivated by psoralen and long-wave UV light.32 PLWUV is known to target nucleic acids preferentially and to introduce chemical cross-links in the viral genome, creating nonreplicating virus. We have confirmed this by titration on BSC40 cells and also by a decrease in protein synthesis in DCs infected with PLWUV-treated rVV. The extent of protein synthesis, as monitored with the VV1-6B6 antibody, is quite variable from one experiment to another, probably because of the random nature with which UV light cross-links the viral genome. The PLWUV-treated rVV retains some toxicity, albeit delayed compared with live rVV (Fig 3A). This has been described for other cells and is not surprising, because ds RNA is still produced.51 52 Also, some of the cytopathic effects are potentially caused by viral enzymes that are brought into the cell with the infectious particles.

However, remarkably, DCs that are infected with PLWUV-treated rVV retain the capacity to present the recombinant EBV antigens. This is also the case with rVV expressing the matrix gene from influenza virus. This finding is reminiscent of prior work in which heat-inactivated or UV-inactivated influenza virus can be efficiently presented by human DCs.53 The current interpretation of these findings is that DCs are such efficient APCs that they are able to elicit immunity with relatively small amounts of viral protein synthesis. The use of PLWUV-treated rVV should make it possible to charge DCs from any individual with EBV antigens without first having to define the peptide that is appropriate for his or her HLA type. Furthermore, PLWUV-treated rVV may have value for immunotherapy of immunosupressed patients, especially if the CD8 repertoire remains intact, or in posttreatment vaccination protocols.

The authors thank Judy Adams and Frank Isdell for assistance with graphics and flow cytometry respectively. We are grateful to Dr B. Moss for recombinant vaccinia matrix virus. We thank Dr C. Münz for critical discussion of the manuscript.

Supported by grants from the National Institutes of Health (AI 40874 to R.M.S. and AI 39516 to N.B.), the Cancer Research Institute (New York, NY), and the National Cancer Institute (to M.G.K.). M.S. was supported by a grant from the Deutsche Forschungsgemeinschaft (DFG).

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

1
Klein
G
Epstein-Barr virus strategy in normal and neoplastic B cells.
Cell
77
1994
791
2
Rickinson
AB
Moss
DJ
Human cytotoxic T lymphocyte responses to Epstein-Barr virus infection.
Annu Rev Immunol
15
1997
405
3
Deeg
HJ
Socié
G
Malignancies after hematopoietic stem cell transplantation: Many questions, some answers.
Blood
91
1998
1833
4
Greenspan
JS
Greenspan
D
Lennette
ET
Abrams
DI
Conant
MA
Petersen
V
Freese
UK
Replication of Epstein-Barr virus within the epithelial cells of oral “hairy” leukoplakia, an AIDS-associated lesion.
N Engl J Med
313
1985
1564
5
Papadopoulos
EB
Ladanyi
M
Mackinnon
S
Boulad
F
Carabasi
MH
Castro-Malaspina
HC
Childs
BH
Gillio
AP
Small
TN
Young
JW
Kernan
NA
O’Reilly
RJ
Infusion of donor leukocytes to treat Epstein-Barr virus-associated lymphoproliferative disorders after allogeneic bone marrow transplantation.
N Engl J Med
330
1994
1185
6
Rooney
CM
Smith
CA
Ng
CYC
Loftin
S
Li
C
Krance
RA
Brenner
MK
Heslop
HE
Use of gene-modified virus-specific T lymphocytes to control Epstein-Barr-virus-related lymphoproliferation.
Lancet
345
1995
9
7
Heslop
HE
Ng
CYC
Li
C
Smith
CA
Loftin
SK
Krance
RA
Brenner
MK
Rooney
CM
Long-term restoration of immunity against Epstein-Barr virus infection by adoptive transfer of gene-modified virus-specific T lymphocytes.
Nat Med
2
1996
551
8
Rooney
CM
Smith
CA
Ng
CYC
Loftin
SK
Sixbey
JW
Gan
Y
Srivastava
D-K
Bowman
LC
Krance
RA
Brenner
MK
Heslop
HE
Infusion of cytotoxic T cells for the prevention and treatment of Epstein-Barr virus-induced lymphoma in allogeneic transplant recipients.
Blood
92
1998
1549
9
Hart
DNJ
Dendritic cells: Unique leukocyte populations which control the primary immune response.
Blood
90
1997
3245
10
Banchereau
J
Steinman
RM
Dendritic cells and the control of immunity.
Nature
392
1998
245
11
Porgador
A
Snyder
D
Gilboa
E
Induction of antitumor immunity using bone marrow-generated dendritic cells.
J Immunol
156
1996
2918
12
Zitvogel
L
Mayordomo
JI
Tjandrawan
T
DeLeo
AB
Clarke
MR
Lotze
MT
Storkus
WJ
Therapy of murine tumors with tumor peptide pulsed dendritic cells: Dependence on T-cells, B7 costimulation, and Th1-associated cytokines.
J Exp Med
183
1996
87
13
Celluzzi
CM
Mayordomo
JI
Storkus
WJ
Lotze
MT
Falo
LD
Jr.
Peptide-pulsed dendritic cells induce antigen-specific, CTL-mediated protective tumor immunity.
J Exp Med
183
1996
283
14
Specht
JM
Wang
G
Do
MT
Lam
JS
Royal
RE
Reeves
ME
Rosenberg
SA
Hwu
P
Dendritic cells retrovirally transduced with a model tumor antigen gene are therapeutically effective against established pulmonary metastases.
J Exp Med
186
1997
1213
15
Song
W
Kong
H
Carpenter
H
Torii
H
Granstein
R
Rafii
S
Moore
MAS
Crystal
RG
Dendritic cells genetically modified with an adenovirus vector encoding the cDNA for a model tumor antigen induce protective and therapeutic antitumor immunity.
J Exp Med
186
1997
1247
16
Mackey
MF
Gunn
JR
Maliszewski
C
Kikutani
H
Noelle
RJ
Barth
RJJ
Dendritic cells require maturation via CD40 to generate protective antitumor immunity.
J Immunol
161
1998
2094
17
DeMatos
P
Abdel-Wahab
Z
Vervaert
C
Seigler
HF
Vaccination with dendritic cells inhibits the growth of hepatic metastases in B6 mice.
Cell Immunol
185
1998
65
18
Fields
RC
Shimizu
K
Mule
JJ
Murine dendritic cells pulsed with whole tumor lysates mediate potent antitumor immune responses in vitro and in vivo.
Proc Natl Acad Sci USA
95
1998
9482
19
Ludewig
B
Ehl
S
Karrer
U
Odermatt
B
Hengartner
H
Zinkernagel
RM
Dendritic cells efficiently induce protective antiviral immunity.
J Virol
272
1998
3812
20
Su
H
Messer
R
Whitmire
W
Fischer
E
Portis
JC
Caldwell
HD
Vaccination against chlamydial genital tract infection following immunization with dendritic cells pulsed ex vivo with non-viable chlamydiae.
J Exp Med
188
1998
809
21
Murray
RJ
Kurilla
MG
Brooks
JM
Thomas
WA
Rowe
M
Kieff
E
Rickinson
AB
Identification of target antigens for the human cytotoxic T cell response to Epstein-Barr virus (EBV): Implications for the immune control of EBV-positive malignancies.
J Exp Med
176
1992
157
22
Steven
NM
Leese
AM
Annels
NE
Lee
SP
Rickinson
AB
Epitope focusing in the primary cytotoxic T cell response to Epstein-Barr virus and its relationship to T cell memory.
J Exp Med
184
1996
1801
23
Callan
MFC
Tan
L
Annels
N
Ogg
GS
Wilson
JDK
O’Callaghan
CA
Steven
N
McMichael
AJ
Rickinson
AB
Direct visualization of antigen-specific CD8+ T cells during the primary immune response to Epstein-Barr virus in vivo.
J Exp Med
187
1998
1395
24
Thomas
JA
Hotchin
N
Allday
MJ
Yacoub
M
Crawford
DH
Immunohistology of Epstein-Barr virus associated antigens in B cell disorders from immunocompromised individuals.
Transplantation
49
1990
944
25
Gratama JW, Zutter MM, Minarovits J, Oosterveer MAP, Thomas ED, Klein G, Ernberg I: Expression of Epstein-Barr virus growth-transformation-associated proteins in lymphoproliferations of bone-marrow transplant recipients. Int J Cancer 188, 1991
26
Murray
RJ
Kurilla
MG
Griffin
HM
Brooks
JM
Mackett
M
Arrand
JR
Rowe
M
Burrows
SR
Moss
DJ
Kieff
E
Rickinson
AB
Human cytotoxic T-cell responses against Epstein-Barr virus nuclear antigens demonstrated by using recombinant vaccinia viruses.
Proc Natl Acad Sci USA
87
1990
2906
27
Bhardwaj
N
Young
JW
Nisanian
AJ
Baggers
J
Steinman
RM
Small amounts of superantigen, when presented on dendritic cells, are sufficient to initiate T cell responses.
J Exp Med
178
1993
633
28
Bender
A
Sapp
M
Schuler
G
Steinman
RM
Bhardwaj
N
Improved methods for the generation of dendritic cells from nonproliferating progenitors in human blood.
J Immunol Methods
196
1996
121
29
Romani
N
Reider
D
Heuer
M
Ebner
S
Eibl
B
Niederwieser
D
Schuler
G
Generation of mature dendritic cells from human blood: An improved method with special regard to clinical applicability.
J Immunol Methods
196
1996
137
30
Reddy
A
Sapp
M
Feldman
M
Subklewe
M
Bhardwaj
N
A monocyte conditioned medium is more effective than defined cytokines in mediating the terminal maturation of human dendritic cells.
Blood
90
1997
3640
31
Smith
GL
Levin
JZ
Palese
P
Moss
B
Synthesis and cellular location of the ten influenza polypeptides individually expressed by recombinant vaccinia viruses.
Virology
160
1987
336
32
Tsung
K
Yim
JH
Marti
W
Buller
RML
Norton
JA
Gene expression and cytopathic effect of vaccinia virus inactivated by psoralen and long-wave UV light.
J Virol
70
1996
165
33
Rowe
M
Evans
HS
Young
LS
Hennessey
K
Kieff
E
Rickinson
AB
Monoclonal antibodies to the latent membrane protein of Epstein-Barr virus reveal heterogeneity of the protein and inducible expression in virus-transformed cells.
J Gen Virol
68
1987
1575
34
Hill
AB
Lee
SP
Haurum
JS
Murray
N
Yao
Q-Y
Rowe
M
Signoret
N
Rickinson
AB
McMichael
AJ
Class I major histocompatibility complex-restricted cytotoxic T lymphocytes specific for Epstein-Barr virus [EBV] nuclear antigens fail to lyse the EBV-transformed B lymphoblastoid cell lines against which they were raised.
J Exp Med
181
1995
2221
35
Shi
Y
Smith
KD
Kurilla
MG
Lutz
CT
Cytotoxic CD8+ T cells recognize EBV antigen but poorly kill autologous EBV-infected B lymphoblasts.
J Immunol
159
1997
1844
36
Apollini
A
Moss
D
Stumm
R
Burrows
S
Suhrbier
A
Misko
I
Schmidt
C
Sculley
T
Sequence variation of cytotoxic T cell epitopes in different isolates of Epstein-Barr virus.
Eur J Immunol
22
1992
183
37
Engelmayer J, Larsson M, Subklewe M, Chahroudi A, Schmaljohn A, William C, Steinman RM, Bhardwaj N: Vaccinia virus inhibits the maturation of human dendritic cells: A novel mechanism of immune evasion. (submitted)
38
Albert
ML
Sauter
B
Bhardwaj
N
Dendritic cells acquire antigen from apoptotic cells and induce class I-restricted CTLs.
Nature
392
1998
86
39
Young
JW
Steinman
RM
Dendritic cells stimulate primary human cytolytic lymphocyte responses in the absence of CD4+ helper T cells.
J Exp Med
171
1990
1315
40
Bhardwaj
N
Bender
A
Gonzalez
N
Bui
LK
Garrett
MC
Steinman
RM
Influenza virus-infected dendritic cells stimulate strong proliferative and cytolytic responses from human CD8+ T cells.
J Clin Invest
94
1994
797
41
Misko
IS
Schmidt
C
Honeyman
M
Soszynski
TD
Sculley
TB
Burrows
SR
Moss
DJ
Burman
K
Failure of Epstein-Barr virus-specific cytotoxic T lymphocytes to lyse B cells transformed with the B95-8 strain is mapped to an epitope that associates with the HLA-B8 antigen.
Clin Exp Immunol
87
1992
65
42
Roskrow
MA
Suzuki
N
Gan
Y-j
Sixbey
JW
Ng
CYC
Kimbrough
S
Hudson
M
Brenner
MK
Heslop
HE
Rooney
CM
Epstein-Barr Virus (EBV)-specific cytotoxic T lymphocytes for the treatment of patients with EBV-positive relapsed Hodgkin’s disease.
Blood
91
1998
2925
43
Walter
EA
Greenberg
PD
Gilbert
MJ
Finch
RJ
Watanabe
KS
Thomas
ED
Riddell
SR
Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor.
N Engl J Med
333
1995
1038
44
Brossart
P
Goldrath
AW
Butz
EA
Martin
S
Bevan
MJ
Virus-mediated delivery of antigenic epitopes into dendritic cells as a means to induce CTL.
J Immunol
158
1997
3270
45
Fan
Z
Huang
XL
Zheng
L
Wilson
C
Borowski
L
Liebmann
J
Gupta
P
Margolick
J
Rinaldo
C
Cultured blood dendritic cells retain HIV-1 antigen-presenting capacity for memory CTL during progressive HIV-1 infection.
J Immunol
159
1997
4973
46
Bronte
V
Carroll
MW
Goletz
TJ
Overwijk
WW
Marincola
F
Rosenberg
SA
Moss
B
Restifo
NP
Antigen expression by dendritic cells correlates with the therapeutic effectiveness of a model recombinant poxvirus tumor vaccine.
Proc Natl Acad Sci USA
94
1997
3183
47
Broder
CC
Kennedy
PE
Michaels
F
Berguer
EA
Expression of foreign genes in cultured human primary macrophages using recombinant vaccinia virus vectors.
Gene
142
1994
167
48
Brown
CK
Turner
PC
Moyer
RW
Molecular characterization of the vaccinia virus hemagglutinin gene.
J Virol
65
1991
3598
49
Niles
EG
Seto
J
Vaccinia virus gene D8 encodes a virion transmembrane protein.
J Virol
62
1988
3772
50
Kibler
KV
Shors
T
Perkins
KB
Zeman
CC
Banaszak
MP
Biesterfeldt
J
Langland
JO
Jacobs
BL
Double-stranded RNA is a trigger for apoptosis in vaccinia virus-infected cells.
J Virol
71
1997
1992
51
Hanafusa
H
Killing of L-cells by heat- and UV-inactivated vaccinia virus.
Biken J
3
1960
191
52
Grunwald-Beard
L
Gamliel
H
Parag
G
Vedantham
S
Zakay-Rones
Z
Killing of Burkitt-lymphoma-derived Daudi cells by ultraviolet-inactivated vaccinia virus.
J Cancer Res Clin Oncol
117
1991
561
53
Bender
A
Bui
LK
Feldman
MAV
Larsson
M
Bhardwaj
N
Inactivated influenza virus, when presented on dendritic cells, elicits human CD8+ cytolytic T cell responses.
J Exp Med
182
1995
1663

Author notes

Address reprint requests to Marion Subklewe, MD, Laboratory of Cellular Physiology and Immunology, Rockefeller University, New York, NY 10021-6399; e-mail: subklem@rockvax.rockefeller.edu.

Sign in via your Institution