We have analyzed paraffin sections from 190 patients with histologically confirmed Hodgkin’s disease (HD) for the presence of Epstein-Barr virus (EBV) using in situ hybridization to detect the EBV-encoded Epstein-Barr virus early RNAs (EBERs) and immunohistochemistry to identify latent membrane protein-1 (LMP1) expression. EBV was present in the tumor cells in 51 HD cases (27%) and was mainly confined to the mixed cellularity and nodular sclerosis subtypes. There was no difference between EBV-positive and EBV-negative HD patients with regard to age, clinical stage, presentation, and the number of alternating chemotherapy cycles of ChIVPP and PABIOE received. The complete remission rate after study chemotherapy was 80% in EBV-positive patients versus 69% in EBV-negative patients (P = .05). The 2-year failure-free survival rate was significantly better for EBV-positive patients when compared with the EBV-negative HD group (P = .02). Although 2-year and 5-year overall survival rates were better for EBV-positive HD patients, the differences were not statistically significant (P = .18 andP = .40, respectively). In conclusion, the results confirm the favorable prognostic value of EBV in the tumor cells of HD patients and suggest important differences in response to chemotherapy between EBV-positive and EBV-negative patients.

THE EPSTEIN-BARR virus (EBV), a B-lymphotropic herpesvirus that infects the majority of the world’s adult population, has been linked to a number of human malignancies, including Burkitt’s lymphoma, nasopharyngeal carcinoma, and Hodgkin’s disease (HD). The observation that persons with a history of infectious mononucleosis have a twofold to threefold increased risk of HD,1 and the detection of elevated levels of antibodies to viral antigens in HD patients before or at the time of diagnosis2-4 provided indirect evidence for a causal role for EBV in HD.

Direct evidence for the association was supplied by the localization of EBV DNA to the malignant Hodgkin and Reed-Sternberg (HRS) cells of HD.5 However, differences in the percentage of EBV-positive HD cases detected in various studies mainly depended on the methods used. Southern blot analysis and in situ hybridization with the use of the BamHI W fragment as a probe yielded between 20% and 40% positive cases.6-8 The highest percentage of positive cases has been obtained using the polymerase chain reaction (PCR) method,9-11 although these studies also detect the presence of EBV infection of bystander reactive lymphocytes and therefore overestimate the EBV association in HD. Perhaps the most widely used and most reliable method is in situ hybridization that targets the highly abundant Epstein-Barr virus early RNAs (EBERs). Using this approach, a variety of studies have demonstrated the presence of latent EBV infection in HRS cells. However, the frequency with which EBV is demonstrated in HD tumors also shows geographical variability, with percentages of between 40% and 50% for North American and European cases,12-15 57% for HD in China,16 but much higher rates in developing countries such as Peru17 and Kenya.18-20 Furthermore, several investigators have demonstrated the clonality of EBV in HD tissue by hybridization with the viral terminal repeat sequences.6,21 These findings indicate clonal expansion of single EBV-infected cells and underline a possible etiological role of EBV in a proportion of HD cases. In addition, immunohistochemical analysis has demonstrated that the HRS cells of EBV-positive cases express high levels of the EBV-encoded latent membrane protein-1 (LMP1).22,23 These data are supported by transcriptional analysis on fresh biopsies of HD that show EBV nuclear antigen-1 (EBNA1) mRNA together with LMP1 and LMP2A and LMP2B transcripts.24 

Although the association between HD and EBV is now established, a potentially more important question is whether the presence of EBV within HRS cells influences outcome for patients with HD. This question has been addressed in three previous studies resulting in contradictory findings.25-27 The first two studies, which comprised relatively small numbers of subjects, failed to identify any effect of EBV on survival.25,26 In contrast, the third study demonstrated a significant association between expression of LMP1 by HRS cells and improved survival in 140 HD patients.27 Also, one of the first two studies used PCR to detect EBV,25 an approach that cannot determine the location of EBV within HD tissues and does not rule out the possible detection of EBV in nonmalignant B lymphocytes.

Therefore, the present study was performed to further examine the prognostic significance of EBV infection in HD, with the use of a clinical trials database that contained detailed information on patient demography, response to chemotherapy, and survival.

Patients

In January 1983, the United Kingdom Central Lymphoma Group (CLG) commenced a study of alternating ChIVPP/PABIOE chemotherapy in previously untreated patients with advanced HD who were unsuitable for treatment with radiotherapy alone. The results of this trial have been reported elsewhere.28 Of the total of 280 patients recruited to this study, paraffin wax-embedded tissue blocks were available for 225 cases.

Histological Review

Hematoxylin and eosin-stained slides were prepared and used to review and subtype all 225 cases according to the Rye classification system. A histological diagnosis of HD was confirmed in 190 of 225 cases. The remaining cases were either not considered to have HD or there was insufficient material remaining in the tissue block to allow a precise histological diagnosis to be made. Before the immunohistochemical and in situ hybridization assays, 4-μm paraffin sections were prepared, deparaffinized, and washed in Tris-buffered saline (TBS), pH 7.6.

Detection of Latent EBV Infection

In situ hybridization for the detection of EBERs was performed according to standard methodology29 and was used to detect the presence of latent EBV infection in all HD samples. Positive controls for EBER in situ hybridization included paraffin wax sections of lymphoblastoid cell lines (LCLs) grown as solid tumors in severe combined immunodeficiency (SCID) mice and a known EBER-positive HD case. U6 and sense control probes were also included in all runs, and their use has been previously described elsewhere.29 Immunohistochemistry for LMP1 was performed on selected cases to confirm the presence of EBV infection in HRS cells. The standard alkaline phosphatase antialkaline phosphatase (APAAP) method was used, and sections were microwave-pretreated, as previously described.30 Positive controls for LMP1 consisted of paraffin wax sections of LCLs grown as solid tumors in SCID mice. Negative controls consisted of consecutive test sections in which primary antibody was replaced with nonimmune serum of the same IgG subclass.

Statistical Methods

Comparison of subgroup with EBV status with those with no EBV status.

Data were collected on 280 HD patients in the trial. EBV status was determined in 190 (68%) of these patients. The aim of the initial analysis was to investigate if the group in whom EBV status had been investigated was representative of all patients entered in the trial.

χ2 tests (including a continuity correction for two-level variables) were used to compare the groups in terms of baseline patient characteristics including gender, prior treatment, histology, symptoms, clinical stage, and all sites of disease (except Waldeyer’s ring for which the Fisher’s exact test was used). Wilcoxon tests were used to compare the groups in terms of age and follow-up, and log-rank tests were used to compare the groups in terms of survival and failure-free survival. Only patients who did not receive previous treatment were included in the investigation of follow-up, overall survival, and failure-free survival.

Comparison of EBV-positive with EBV-negative trial patients.

The second part of the analysis was used only for those patients in whom EBV status was known and involved a comparison of EBV-negative and EBV-positive groups. A combination of χ2 tests, Fisher’s exact tests (both for categorical data), and Wilcoxon tests (continuous data) were used to compare these groups in terms of gender, age, prior treatment, histology, symptoms, clinical stage, sites of disease, and follow-up. Survival and failure-free survival were compared with the use of log-rank tests and Kaplan-Meier curves. Survival was calculated as the time from beginning chemotherapy to date of death from any cause or date of the last known follow-up. For those patients who responded to treatment, failure-free survival was calculated as the time from beginning chemotherapy to either the date of relapse, or in those patients with no previous documented relapse, either the date of death from any cause or date of the last known follow-up. For those patients who did not respond to chemotherapy, failure-free survival was recorded as zero.

Detection of Latent EBV Infection in HD Samples

Of the 190 HD patients, the presence of latent EBV infection, as detected by in situ hybridization for the EBERs, was identified in HRS cells in 51 (27%) cases (designated as the EBV-positive group). The remaining 139 (73%) cases did not show the presence of the EBERs within HRS cells and will subsequently be referred to as the EBV-negative group. Immunohistochemistry for the LMP1 protein gave similar results to EBER in situ hybridization.

Comparison of Subgroup With EBV Status

When the group with known EBV status (n = 190) was compared with the individuals with no EBV status (n = 90), there was found to be no significant difference in terms of any of the parameters studied with the exception of prior treatment (P = .03). Because those who had prior treatment were already excluded from any analysis of outcome variables, this difference is not relevant.

Comparison of EBV-Positive and EBV-Negative Patients

Patient characteristics.

The results showed that there was no significant difference between the EBV-negative and EBV-positive groups in terms of clinical stage or site(s) of disease at presentation, age, or presence of B symptoms (Table 1). However, a significantly greater proportion of EBV-positive patients were male compared with the EBV-negative group (P = .001). Even when the P values were adjusted, with the use of Bonferroni’s correction to account for multiple characteristics being tested, gender remained significant at the 5% level (P = .01).

Table 1.

Characteristics of 190 HD Patients With Known EBV Status

Characteristic EBVPositive EBVNegativeP Value
Gender: male  43 (84%)  80 (58%) .001  
Median age (inter-quartile range)  33 (22-49) 33 (23-45)  .55  
B symptoms: present  16 (31%) 53 (38%)  .49  
Clinical stage  
 1  2 (4%) 10 (7%)  .87  
 2  16 (33%)  45 (33%)  
 3 19 (39%)  48 (35%)  
 4  12 (24%)  35 (25%) 
Histological type  
 Lymphocyte predominant  2 (5%) 11 (9%)  
 Nodular sclerosis (NS)  30 (68%) 86 (69%)  1.00  
 Mixed cellularity  10 (23%) 17 (14%)  
 Lymphocyte depleted (LD)  1 (2%)  5 (4%) 
 NS + LD  1 (2%)  5 (4%)  
 NK  7  12 
Enlarged lymph nodes  
 Cervical  32 (63%) 100 (72%)  .30  
 Axillary  11 (22%)  49 (35%) .11  
 Inguinal  7 (14%)  23 (17%)  .80 
 Abdominal  9 (18%)  25 (18%)  1.00  
 Waldeyer’s ring  3 (2%)  1 (1%)  1.00  
Hepatomegaly  4 (8%) 14 (10%)  .78  
Splenomegaly  13 (25%)  22 (16%) .20  
Total no. of patients (n)  51  139 
Characteristic EBVPositive EBVNegativeP Value
Gender: male  43 (84%)  80 (58%) .001  
Median age (inter-quartile range)  33 (22-49) 33 (23-45)  .55  
B symptoms: present  16 (31%) 53 (38%)  .49  
Clinical stage  
 1  2 (4%) 10 (7%)  .87  
 2  16 (33%)  45 (33%)  
 3 19 (39%)  48 (35%)  
 4  12 (24%)  35 (25%) 
Histological type  
 Lymphocyte predominant  2 (5%) 11 (9%)  
 Nodular sclerosis (NS)  30 (68%) 86 (69%)  1.00  
 Mixed cellularity  10 (23%) 17 (14%)  
 Lymphocyte depleted (LD)  1 (2%)  5 (4%) 
 NS + LD  1 (2%)  5 (4%)  
 NK  7  12 
Enlarged lymph nodes  
 Cervical  32 (63%) 100 (72%)  .30  
 Axillary  11 (22%)  49 (35%) .11  
 Inguinal  7 (14%)  23 (17%)  .80 
 Abdominal  9 (18%)  25 (18%)  1.00  
 Waldeyer’s ring  3 (2%)  1 (1%)  1.00  
Hepatomegaly  4 (8%) 14 (10%)  .78  
Splenomegaly  13 (25%)  22 (16%) .20  
Total no. of patients (n)  51  139 

Abbreviation: NK, not known.

Treatment outcome.

Ten patients from the EBV-positive group and 19 from the EBV-negative group had received prior treatment, and these were eliminated from the analysis at this stage. Median follow-up for the 123 patients still alive in the eligible group was 86 months (interquartile range, 57 to 107 months). There were no significant differences in follow-up for the different EBV status groups (P = .99). χ2 tests also showed that there were no statistically significant differences between the groups in terms of the numbers of chemotherapy cycles received (P = .48 for ChIVPP and P = .69 for PABIOE).

When χ2 tests were used to compare groups in terms of response to treatment (Table2), it was shown that EBV-positive patients were more likely to achieve a complete response to chemotherapy than EBV-negative patients (P = .05).

Table 2.

Effect of EBV Infection of HRS Cells on Treatment Outcome and Survival in HD Patients

EBVPositive EBVNegative P Value
Side effects during therapy  
 Hematological toxicity 18 (44%)  57 (48%)  .79  
 Infection 17 (41%)  60 (50%)  .42  
 Neuropathy  9 (22%)  21 (18%)  .71  
Response 
 CR  33 (80%)  82 (69%)  .05  
 PR  4 (10%)  31 (26%)  
 Fail  4 (10%)   5 (4%)   
 NK  0  
Survival  
 2-yr survival rate  90.2%  81.7% 
  Difference (95% CI)   8.5% (−3% to 20%)  .18 
 5-yr survival rate  84.5%  78.2%  
  Difference (95% CI)   6.3% (−7.9% to 20.5%)  .40  
 2-yr failure-free survival  90.2%  74.2%  
  Difference (95% CI)   16% (−3.9%-28.1%)  .02  
 5-yr failure-free survival  81.5%  68.5%  
  Difference (95% CI)   13% (−2.9% to 28.8%)  .13 
EBVPositive EBVNegative P Value
Side effects during therapy  
 Hematological toxicity 18 (44%)  57 (48%)  .79  
 Infection 17 (41%)  60 (50%)  .42  
 Neuropathy  9 (22%)  21 (18%)  .71  
Response 
 CR  33 (80%)  82 (69%)  .05  
 PR  4 (10%)  31 (26%)  
 Fail  4 (10%)   5 (4%)   
 NK  0  
Survival  
 2-yr survival rate  90.2%  81.7% 
  Difference (95% CI)   8.5% (−3% to 20%)  .18 
 5-yr survival rate  84.5%  78.2%  
  Difference (95% CI)   6.3% (−7.9% to 20.5%)  .40  
 2-yr failure-free survival  90.2%  74.2%  
  Difference (95% CI)   16% (−3.9%-28.1%)  .02  
 5-yr failure-free survival  81.5%  68.5%  
  Difference (95% CI)   13% (−2.9% to 28.8%)  .13 

Abbreviations: CR, complete response; PR, partial response; NK, not known; CI, confidence interval.

EBV-positive HD patients had longer 2-year and 5-year overall survival rates (Fig 1) than the EBV-negative group, but these differences were not statistically significant (P = .18 and P = .40, respectively; Table 2). However, 2-year failure-free survival rates were significantly better for EBV-positive versus EBV-negative HD patients (P = .02; Fig 2 and Table 2). Similarly, the 5-year failure-free survival rate was better for EBV-positive patients, but this difference was not statistically significant (P = .13; Fig 2 and Table 2).

Fig. 1.

Survival curves by EBV status.

Fig. 1.

Survival curves by EBV status.

Close modal
Fig. 2.

Failure-free survival by EBV status.

Fig. 2.

Failure-free survival by EBV status.

Close modal

There is accumulating evidence to suggest that EBV is not merely a silent passenger in HD but plays a key role in its pathogenesis. This is largely based on the demonstration of monoclonal EBV episomes in HD tumors,6,21 implying EBV infection as an initiating event and on the high level expression of the EBV-encoded LMP1 by HRS cells.22-24 

However, few studies have focused on the potential prognostic significance of EBV in HD. On theoretical grounds, there are two opposing biological effects that could impact on prognosis. Firstly, by virtue of its well-documented effects promoting cell growth in vitro, the expression of LMP1 in the malignant cells of HD might be expected to exert a negative effect on survival for EBV-positive patients. LMP1 has been shown to have transforming effects in rodent fibroblast cell lines,31,32 enabling Rat-1 or NIH 3T3 cells to grow in medium supplemented with low serum.31 LMP1 also induces loss of contact inhibition in Rat-1 cells and causes both Rat-1 and BALB/c 3T3 cells to lose their anchorage dependence so that they clone with high efficiency in soft agar.31-33 Rat-1 cells expressing LMP1 are also tumorigenic in nude mice, whereas control Rat-1 cells are not.31 

On the other hand, cytotoxic T lymphocyte (CTL) responses to LMP1 and LMP2, proteins that are consistently expressed by EBV-positive HRS cells, have been demonstrated in the blood of EBV-seropositive individuals.34-38 Furthermore, LMP2-specific CTLs have been identified and expanded from the peripheral blood of HD patients,39,40 suggesting that expression of LMP2 by HRS cells might stimulate an antineoplastic CTL response. Recent studies have also shown that the HD cell line, HDLM2, is able to process and present epitopes from LMP1 and LMP2 and is sensitive to lysis by EBV-specific CTLs.39 LMP1 expression may also increase immune recognition of infected cells in other ways. LMP1 has been shown to upregulate leukocyte function antigen 3 (LFA3) and intercellular adhesion molecule 1 (ICAM1) protein levels41-44 and major histocompatibility complex (MHC) class I and class II expression.45-47 Transfection of BL cells with LMP1 restores their ability to process both endogenous and exogenous antigens and present them to responder T cells.48,49 These observations are supported by the frequent detection of high level expression of human leukocyte antigen (HLA) class I molecules on HRS cells from EBV-positive, but not EBV-negative, HD tumors.50,51 The apparent downregulation of HLA class I molecules on EBV-negative HRS cells may also contribute to poorer survival for EBV-negative HD patients. HLA class I loss has previously been associated with poor prognosis in non-Hodgkin’s lymphomas and in other tumors.52 

A previous study has reported a significant association between the detection of LMP1 in HRS cells and longer survival in a series of 140 HD patients.27 Therefore, both the present study and the Spanish study confirm the favorable prognostic effect of EBV expression in HD. The discrepancy regarding complete remission rates could be because of the different chemotherapy regimens administered: MOPP/ABVD in the Spanish study versus ChIVPP/PABIOE in the present study. Also, the present study excluded children, whereas the Spanish study included both children and adults. Clearly, further study on a larger sample of patients or meta-analysis of existing studies is indicated, not only to increase the power to detect differences in survival between EBV-positive and EBV-negative groups, but also to account for differences in outcome based on subtype association.

The finding that EBV-positive patients were more likely to achieve a complete response to chemotherapy than were EBV-negative patients, suggests that the malignant cells of EBV-positive HD may be more sensitive to the chemotherapy agents or that the residual EBV-positive cells after cytoreduction by chemotherapy might be targets for immune cytolysis. Several recent data indirectly support the hypothesis that the sensitivity of HRS cells to chemotherapy-induced apoptosis is higher in EBV-positive cells. Expression of the anti-apoptotic gene, Bcl-2, by HRS cells has been shown to predict poor clinical outcome in HD patients,53 whereas recent studies have shown that fewer HRS cells of EBV-positive HD express the Bcl-2 gene than do EBV-negative cases.54 Although it is possible that protection from chemotherapy-induced apoptosis in EBV-infected HRS cells may be mediated by other members of the bcl-2 family, including Bcl-x,55 more work is required to establish any effect of levels of expression of these genes on response to chemotherapy.

The demonstration of EBV in the tumors of HD patients may have important implications for other treatment strategies. In particular, there is growing interest in the potential development of immunotherapy approaches that use EBV-specific CTLs to target the EBV-infected malignant cell population. Such an approach has already been successful in the treatment of EBV-associated lymphoproliferative disease where adoptive transfer of donor EBV-specific CTLs has resulted in the eradication of tumors.56,57 The development of this form of therapy for HD may be particularly relevant to certain patients with recurrent or refractory disease, in whom conventional forms of treatment have produced long-term disease-free survival rates of less than 30%.58 

The authors are grateful to Dr M. Cullen of the United Kingdom Central Lymphoma Group for access to patient data used in this study.

Supported by the Cancer Research Campaign.

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
Gutensohn
N
Cole
P
Epidemiology of Hodgkin’s disease.
Semin Oncol
7
1980
92
2
Mueller
N
Evans
A
Harris
NL
Comstock
GW
Jellum
E
Magnus
K
Orentreich
N
Polk
BF
Vogelman
J
Hodgkin’s disease and Epstein-Barr virus. Altered antibody pattern before diagnosis.
N Engl J Med
32
1989
689
3
Levine
PH
Pallesen
G
Ebbesen
P
Harris
N
Evans
AS
Mueller
N
Evaluation of Epstein-Barr virus antibody patterns and detection of viral markers in the biopsies of patients with Hodgkin’s disease.
Int J Cancer
59
1994
48
4
Alexander
FE
Daniel
CP
Armstrong
AA
Clark
DA
Onions
DE
Cartwright
RA
Jarrett
RF
Case clustering, Epstein-Barr virus Reed-Sternberg cell status and herpes virus serology in Hodgkin’s disease: Results of a case control study.
Eur J Cancer
31A
1995
1479
5
Weiss
LM
Strickler
JG
Warnke
RA
Purtilo
DT
Sklar
J
Epstein-Barr viral DNA in tissues of Hodgkin’s disease.
Am J Pathol
129
1987
86
6
Anagnostopoulos
I
Herbst
H
Niedobitek
G
Stein
H
Demonstration of monoclonal EBV genomes in Hodgkin’s disease and Ki-1-positive anaplastic large cell lymphoma by combined Southern blot and in situ hybridization.
Blood
74
1989
810
7
Staal
SP
Ambinder
R
Beschorner
WE
Hayward
GS
Mann
R
A survey of Epstein-Barr virus DNA in lymphoid tissue: Frequent detection in Hodgkin’s disease.
Am J Clin Pathol
91
1989
1
8
Jarrett
RF
Gallagher
A
Jones
DB
Alexander
FE
Krajewski
AS
Kelsey
A
Adams
J
Angus
B
Gledhill
S
Wright
DH
Detection of EBV genomes in Hodgkin’s disease: Relation to age.
J Clin Pathol
44
1991
844
9
Knecht
H
Odermatt
B
Bachmann
E
Teixera
S
Sahli
R
Hayoz
D
Heitz
P
Bachmann
F
Frequent detection of Epstein-Barr virus DNA by the polymerase chain reaction in lymph node biopsies from patients with Hodgkin’s disease without genomic evidence of B- or T-cell clonality.
Blood
78
1991
760
10
Bignon
YJ
Bernard
D
Cure
H
Fonck
Y
Pauchard
J
Travade
P
Legros
M
Dastugue
B
Plagne
R
Detection of Epstein-Barr viral genomes in lymph nodes of Hodgkin’s disease patients.
Mol Carcinog
3
1990
9
11
Herbst
H
Niedobitek
G
Kneba
M
Hummel
M
Finn
T
Anagnostopolous
I
Bergholz
M
Kreiger
C
Stein
H
High incidence of Epstein-Barr virus genomes in Hodgkin’s disease.
Am J Pathol
137
1990
13
12
Wu
TC
Mann
RB
Charache
P
Hayward
SD
Staal
S
Lambe
BC
Ambinder
RF
Detection of EBV gene expression in Reed-Sternberg cells of Hodgkin’s disease.
Int J Cancer
46
1990
801
13
Weiss
LM
Chen
YY
Liu
XF
Shibata
D
Epstein-Barr virus and Hodgkin’s disease: A correlative in situ hybridization and polymerase chain reaction study.
Am J Pathol
139
1991
1259
14
Herbst
H
Steinbrecher
E
Niedobitek
G
Distribution and phenotype of Epstein-Barr virus-harboring cells in Hodgkin’s disease.
Blood
80
1992
484
15
Hummel
M
Anagnostopoulos
I
Dallenbach
F
Korbjuhn
P
Dimmler
C
Stein
H
EBV infection patterns in HD and normal lymphoid tissue: Expression and cellular localization of gene products.
Br J Haematol
82
1992
689
16
Zhu
ZG
Hamilton-Dutoit
S
Yan
QH
Pallesen
G
The association between Epstein-Barr virus and Chinese Hodgkin’s disease.
Int J Cancer
55
1993
359
17
Chang
KL
Albujar
PF
Chen
Y-Y
Johnson
RM
Weiss
LM
High prevalence of Epstein-Barr virus in Reed-Sternberg cells of Hodgkin’s disease occurring in Peru.
Blood
81
1993
496
18
Leoncini
L
Spina
D
Nyong’o
A
Abinya
O
Minacci
C
Disanto
A
De Luca
F
De Vivo
A
Sabattini
E
Poggi
S
Pileri
S
Tosi
P
Neoplastic cells of Hodgkin’s disease show differences in EBV expression between Kenya and Italy.
Int J Cancer
65
1996
781
19
Weinreb
M
Day
PJR
Niggli
F
Green
EK
Nyongo’o
AO
Othieno-Abinya
NA
Riyat
MS
Rafaat
F
Mann
JR
The consistent association between Epstein-Barr virus and Hodgkin’s disease in children in Kenya.
Blood
87
1996
3828
20
Weinreb
M
Day
PJR
Niggli
F
Powell
JE
Raafat
F
Hesseling
PB
Schneider
JW
Hartely
PS
The role of Epstein-Barr virus in Hodgkin’s disease from different geographical areas.
Arch Dis Child
74
1996
27
21
Weiss
LM
Movahed
LA
Warnke
RA
Sklar
J
Detection of Epstein-Barr viral genomes in Reed-Sternberg cells of Hodgkin’s disease.
N Engl J Med
320
1989
502
22
Pallesen
G
Hamilton-Dutoit
SJ
Rowe
M
Young
LS
Expression of Epstein-Barr virus latent gene products in tumor cells of Hodgkin’s disease.
Lancet
337
1991
320
23
Murray
PG
Young
LS
Rowe
M
Crocker
J
Immunohistochemical demonstration of the Epstein-Barr virus-encoded latent membrane protein in paraffin sections of Hodgkin’s disease.
J Pathol
166
1992
1
24
Deacon
EM
Pallesen
G
Niedobitek
G
Crocker
J
Brooks
L
Rickinson
AB
Young
LS
Epstein-Barr virus and Hodgkin’s disease: Transcriptional analysis of virus latency in the malignant cells.
J Exp Med
177
1993
339
25
Fellbaum
C
Hansmann
M-L
Niedermeyer
H
Kraus
I
Alavaikko
MJ
Busch
R
Putz
B
Fischer
R
Hofler
H
Influence of Epstein-Barr virus genomes on patient survival in Hodgkin’s disease.
Am J Clin Pathol
98
1992
319
26
Claviez
A
Tiemann
M
Peters
J
Kreipe
H
Schneppenheim
R
Parwaresch
R
The impact of EBV, proliferation rate and bcl-2 expression in Hodgkin’s disease in childhood.
Ann Hematol
68
1994
6
27
Morente
MM
Piris
MA
Abraira
V
Acevedo
A
Aguilera
B
Bellas
C
Fraga
M
Garcia
R
Gomez-Marcos
F
Menarguez
J
Oliva
H
Sanchez-Beato
M
Montalban
C
Adverse clinical outcome in Hodgkin’s disease is associated with loss of retinoblastoma protein expression, High Ki67 proliferation index and absence of Epstein-Barr virus-latent membrane protein 1 expression.
Blood
90
1997
2429
28
Cullen
MH
Stuart
NSA
Woodroffe
C
Murphy
A
Fletcher
J
Blackledge
GRP
Child
JA
Grieve
RJ
Jones
EL
ChIVPP/PABIOE and radiotherapy in advanced Hodgkin’s disease.
J Clin Oncol
12
1994
779
29
Barletta
JM
Kingma
DW
Charache
P
Mann
RB
Ambinder
RF
Rapid in situ hybridization for the diagnosis of latent Epstein-Barr virus infection.
Mol Cell Probes
7
1993
105
30
Murray
PG
Swinnen
LJ
Constandinou
CM
Pyle
JM
Carr
TJ
Hardwick
JM
Ambinder
RF
bcl-2 but not the EBV-encoded bcl-2 homologue, BHRF1, is commonly expressed in post-transplantation lymphoproliferative disorders.
Blood
87
1996
706
31
Wang
D
Liebowitz
D
Kieff
E
An EBV membrane protein expressed in immortalized lymphocytes transforms established rodent cells.
Cell
34
1985
831
32
Baichwal
VR
Sugden
B
Transformation of Balb/3T3 cells by the BNLF-1 gene of Epstein-Barr virus.
Oncogene
2
1988
461
33
Wang
D
Liebowitz
D
Kieff
E
The truncated form of the Epstein-Barr virus latent-infection membrane protein expressed in virus replication does not transform rodent fibroblasts.
J Virol
62
1988
2337
34
Gavioli
R
Kurilla
M
de Campos-Lima
PO
Wallace
LE
Dolcetti
R
Murray
RJ
Rickinson
AB
Masucci
M
Multiple HLA A11 restricted CTL epitopes of different immunogenicities in the Epstein-Barr virus encoded nuclear antigen-4 (EBNA4).
J Virol
67
1993
1572
35
de Campos-Lima
PO
Gavioli
R
Zhang
QJ
Wallace
L
Rowe
M
Rickinson
AB
Masucci
MG
HLA-A 11 epitope loss isolates of Epstein-Barr virus from a highly A11+ population.
Science
260
1993
98
36
de Campos-Lima
PO
Levitsky
V
Brooks
J
Lee
SP
Hu
LF
Rickinson
AB
Masucci
MG
T cell responses and virus evolution: Epstein-Barr virus isolates from highly HLA A11-positive populations carry mutations selectively involving anchor residues of A11-restricted CTL epitopes.
J Exp Med
179
1994
1297
37
Khanna
R
Burrows
SR
Steigerwald-Mullen
PM
Thomson
SA
Kurilla
M
Moss
DJ
Isolation of cytotoxic T-lymphocytes from healthy seropositive individuals specific for peptide epitopes from Epstein-Barr virus nuclear antigen 1: Implications for viral persistence and tumor surveillance.
Virology
214
1995
633
38
Levitsky
V
Zhang
QJ
Levitskaya
J
Masucci
M
The life span of major histocompatibility complex-peptide complexes influences the efficiency of presentation and immunogenicity of two class I-restricted cytotoxic T-lymphocyte epitopes in the Epstein-Barr virus nuclear antigen 4.
J Exp Med
183
1996
915
39
Sing
A
Ambinder
RF
Hong
DJ
Jensen
M
Batten
W
Petersdorf
E
Greenberg
PD
Isolation of Epstein-Barr virus (EBV)-specific cytotoxic T lymphocytes that lyse Reed-Sternberg cells: Implications for immune-mediated therapy for Hodgkin’s disease.
Blood
89
1997
1978
40
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
41
Wang
F
Gregory
C
Sample
C
Murray
R
Liebowitz
D
Rowe
M
Rickinson
A
Kieff
E
Epstein-Barr virus latent infection membrane and nuclear proteins 2 and 3C are effectors of phenotypic changes in B lymphocytes: EBNA2 and LMP co-operatively induce CD23.
J Virol
64
1990
2309
42
Cuomo
L
Trivedi
P
Wang
F
Winberg
G
Klein
G
Masucci
M
Expression of the Epstein-Barr virus (EBV)-encoded membrane antigen (LMP) increases the stimulatory capacity of EBV-negative B lymphoma lines in allogeneic mixed lymphocyte cultures.
Eur J Immunol
20
1990
2293
43
Peng
M
Lundgren
E
Transient expression of the Epstein-Barr virus LMP1 gene in human primary B cells induces cellular activation and DNA synthesis.
Oncogene
7
1992
1775
44
Rowe
M
Peng-Pilon
M
Huen
DS
Hardy
R
Croom-Carter
D
Lundgren
E
Rickinson
AB
Upregulation of bcl-2 by the Epstein-Barr virus latent membrane protein LMP1: A B-cell-specific response that is delayed relative to NF-κB activation and to induction of cell surface markers.
J Virol
6
1994
5602
45
de Campos-Lima
PO
Torsteinsdottir
S
Cuomo
L
Klein
G
Sulitzeanu
D
Masucci
MG
Antigen processing and presentation by EBV-carrying cell lines: Cell-phenotype dependence and influence of the EBV-encoded LMP1.
Int J Cancer
53
1993
856
46
Zhang
Q
Brooks
L
Busson
P
Wang
F
Charron
D
Kieff
E
Rickinson
AB
Turz
T
Epstein-Barr virus latent membrane-protein-1 increases HLA class-II expression in an EBV-negative B-cell line.
Eur J Immunol
24
1994
1467
47
Rowe
M
Khanna
R
Jacob
CA
Argaet
V
Kelly
A
Powis
S
Bellich
M
Croom-Carter
D
Lee
S
Burrows
SR
Restoration of endogenous antigen processing in Burkitt’s lymphoma cells by Epstein-Barr virus latent membrane protein-1: Coordinate upregulation of peptide transporters and HLA class I antigen expression.
Eur J Immunol
25
1995
1374
48
Longnecker
R
Kieff
E
A second Epstein-Barr virus membrane protein (LMP2) is expressed in latent infection and colocalizes with LMP1.
J Virol
64
1990
2319
49
Rowe
DT
Hall
L
Joab
I
Laux
G
Identification of the Epstein-Barr virus terminal protein gene products in latently infected lymphocytes.
J Virol
64
1990
2866
50
Oudejans
JJ
Jiwa
NM
Kummer
JA
Horstman
A
Vos
W
Baak
JPA
Kluin Ph
M
van der Valk
P
Walboomers
JMM
Meijer
CJLM
Analysis of major histocompatibility complex class I expression on Reed-Sternberg cells in relation to the cytotoxic T cell response in Epstein-Barr virus-positive and -negative Hodgkin’s disease.
Blood
87
1996
3844
51
Murray
PG
Constandinou
CM
Crocker
J
Young
LS
Ambinder
RF
Analysis of MHC class I and TAP expression and LMP2 epitope sequence in EBV-positive Hodgkin’s disease.
Blood
92
1998
2477
52
Amiot
L
Onno
M
Lamy
T
Dauric
C
Le Prise
P
Fauchet
R
Loss of HLA molecules in B lymphomas is associated with an aggressive clinical course.
Br J Haematol
100
1998
655
53
Brink
AA
Oudejans
JJ
van den Brule
AJ
Kluin
PM
Horstman
A
Ossenkoppele
GJ
van Heerde
P
Jiwa
M
Meijer
CJ
Low p53 and high Bcl-2 expression in Reed-Sternberg cells predicts poor clinical outcome for Hodgkin’s disease: Involvement of apoptosis resistance?
Mod Pathol
11
1998
376
54
Jiwa
NM
Oudejans
JJ
Bai
MC
van den Brule
AJ
Horstman
A
Vos
A
van der Valk
P
Kluin
PM
Walboomers
JM
Meijer
CJ
Expression of bcl-2 protein and transcription of the Epstein-Barr virus homologue BHRF-1 in Hodgkin’s disease: Implications for different pathogenic mechanisms.
Histopathology
26
1995
547
55
Schlaifer
D
March
M
Krajewski
S
Laurent
G
Pris
J
Delsol
G
Reed
JC
Brousset
P
High level expression of the bcl-x gene in Reed-Sternberg cells of Hodgkin’s disease.
Blood
85
1995
2671
56
Papadopoulos
EB
Ladanyi
M
Emanuel
D
Mackinnon
S
Boulad
F
Carabasi
MH
Castro-Malaspina
H
Childs
BH
Gillio
AP
Small
TN
Young
JW
Kernan
NA
O’Rielly
RJ
Infusion of donor leukocytes to treat Epstein-Barr virus associated lymphoproliferative disorders after allogeneic bone marrow transplantation.
N Engl J Med
331
1994
679
57
Rooney
CM
Smith
CA
Ng
CYC
Loftin
S
Li
C
Krance
MK
Brenner
MK
Heslop
HE
Use of gene-modified virus-specific T lymphocytes to control Epstein-Barr virus-related lymphoproliferation.
Lancet
345
1995
9
58
Longo
DL
Duffey
PL
Young
RC
Hubbard
SM
Ihde
DC
Glastein
E
Phares
JC
Jaffe
ES
Urba
WJ
DeVita
VT
Jr
Conventional-dose salvage combination chemotherapy in patients relapsing with Hodgkin’s disease after combination chemotherapy: The low probability for cure.
J Clin Oncol
10
1992
210

Author notes

Address reprint requests to Paul G. Murray, PhD, Biomedical Research Laboratories, School of Health Sciences, University of Wolverhampton, Wolverhampton, WV1 1DJ, UK; e-mail:p.g.murray@wlv.ac.uk.

Sign in via your Institution