The Epstein-Barr virus (EBV)-encoded latent membrane proteins, LMP1 and LMP2, are consistently expressed by the malignant Hodgkin/Reed-Sternberg (HRS) cells of EBV-associated Hodgkin's disease (HD). Cytotoxic T lymphocyte (CTL) responses to both of these proteins have been shown in the blood of EBV-seropositive individuals, yet in HD the apparent failure of the CTL response to eliminate HRS cells expressing LMP1 and LMP2 in vivo has given rise to the suggestion that HD may be characterized by the presence of defects in antigen processing/presentation or in CTL function. This study has used immunohistochemistry to show high-level expression of major histocompatibility complex (MHC) class I molecules by the HRS cells of EBV-associated HD and either low level or absence of expression of MHC class I molecules on HRS cells of EBV-negative tumors. In addition, HRS cells expressed high levels of transporter-associated proteins (TAP-1, -2), irrespective of the presence of latent EBV infection. These results suggest that global downregulation of MHC class I molecules does not account for the apparent ability of EBV-infected HRS cells to evade CTL responses, but may be important in the understanding of EBV-negative disease.We have also sequenced an epitope in LMP2A (CLGGLLTMV) that is restricted through HLA A2.1, a relatively common allele in Caucasian populations, and showed that this epitope is wild type in a small group of EBV-associated HLA A2.1-positive HD tumors. This result may be relevant to proposed immunotherapeutic approaches for EBV-positive HD patients that target CTL epitopes.

THE EPSTEIN-BARR VIRUS (EBV) is a human herpesvirus that infects approximately 95% of the world's adult population.1 EBV is generally carried as a clinically silent infection but is strongly implicated in the pathogenesis of a number of human malignancies including African Burkitt's lymphoma,2,3 nasopharyngeal carcinoma,4,5 and more recently, Hodgkin's disease (HD). EBV DNA has been detected in the tumor samples of HD patients by Southern blotting and polymerase chain reaction (PCR) analysis6-9 and a variety of studies have localized viral DNA10 and virus gene products11-13 to the malignant Hodgkin/Reed-Sternberg (HRS) cells of HD. Furthermore, using EBV terminal repeat probes, the EBV genomes within individual cases have been shown to be monoclonal in origin, suggesting that monoclonal proliferation occurred after EBV infection.7,8,10,14 The frequent occurrence of abnormally elevated titers of antibodies against EBV antigens before the onset of overt disease and the detection of impaired cellular responses to EBV in HD patients have led to the hypothesis that a defective control of EBV-infected cells might be important in the pathogenesis of a proportion of HD tumors.15-19 EBV-infected HRS cells are characterized by a pattern of viral protein expression that is restricted to EBNA1, LMP1, and LMP2.12,20-23 Although the dominant CTL responses seem to be directed to epitopes in proteins of the EBNA3 family, LMP1 and LMP2 have been shown to be targets for autologous cytotoxic T lymphocytes (CTLs) in vitro.24-27The apparent failure of the immune system to eliminate HRS cells expressing potential targets for CTLs has provided further support for immunologic dysfunction in EBV-associated HD.

One possible mechanism of CTL escape in EBV-associated HD may be downregulation of major histocompatibility complex (MHC) class I molecules on HRS cells. CTLs recognize endogenously processed virus proteins only when presented at the cell surface as peptides bound to MHC class I molecules. A reduction or the absence of MHC class I expression can therefore render virus-infected cells resistant to CTL destruction. Such a mechanism has been shown previously for some virus-associated tumors, including EBV-associated Burkitt's lymphoma (BL) and a proportion of human papilloma virus (HPV)-infected cervical carcinomas.28-31 In the case of HPV-positive cervical carcinomas, MHC class I downregulation was associated with the loss of function of one of the transporter-associated proteins (TAP-1).32,33 

It has been shown that amino acid substitutions at key positions within peptides presented by MHC class I molecules may reduce or completely abrogate CTL responses. For example, two HLA A11-restricted CTL epitopes (residues 399-408 and 416-424) from the EBNA3B protein are regularly mutated in EBV strains from Southeast Asia,34,35and memory CTL responses specific for the variant epitopes have not been detectable in HLA A11-positive Chinese donors infected with the mutated viruses. It is possible that similar mechanisms might also account for the failure of CTLs to eliminate EBV-infected HRS cells.

Therefore, the aim of this current study was to investigate the expression of MHC class I and TAP molecules in both EBV-positive and EBV-negative HD. The identification of an epitope in LMP2(CLGGLLTMV) that is restricted through HLA A2.1, a relatively common allele in Caucasian populations, provides an ideal target to study the possibility that epitope variations in EBV genes expressed in tumor tissues might be responsible for CTL escape.36 

Preparation of specimens.

Paraffin-wax sections and paraffin-wax–embedded tissue blocks from 39 cases of HD were available from the pathology files of the Johns Hopkins Hospital, Baltimore, MD; and the Queen Elizabeth Hospital, Birmingham, UK. From tissue blocks 4-μm paraffin sections were prepared and all sections were deparaffinized and washed in Tris-buffered saline (TBS), pH 7.6. Cryostat sections were also prepared from selected samples. Hematoxylin and eosin–stained sections of HD tumors were reviewed and subtyped according to the Rye classification system.

Detection of latent EBV infection.

In situ hybridization for the detection of the Epstein-Barr virus early RNAs (EBERs) was performed according to standard methods.37Positive controls for EBER in situ hybridization included paraffin-wax sections of lymphoblastoid cell lines (LCL) grown as solid tumors in severe combined immunodeficiency (SCID) mice (LCL-SCID tumors) and a known EBER-positive HD case. U6 and sense control probes were included in all runs.

Immunohistology.

Immunohistochemistry for LMP1 and LMP2 was also performed on selected cases to confirm the presence of latent EBV infection in HRS cells. Immunostaining for MHC class I and TAP-1 was performed on acetone-fixed frozen and paraffin-wax sections of HD tissues and on a variety of cell lines (Table 1), including the HD lines HDLM2, L428, HS445, and Km-H2. HD cell lines and selected HD biopsies were also stained for TAP-2. Immunohistochemistry for TAP-1 and TAP-2 was additionally performed on acetone-fixed cytospin preparations of the BL lines, Mutu I (group I BL) and Mutu III (group III BL).

Table 1.

Cell Lines Used in the Study

Cell Line Comments
B95.859 Marmoset cell line 
X50-760 Cord blood LCL  
Coneron LCL* HLA A2.1 homozygous  
Bevan LCL* HLA A2.1 homozygous  
M Rowe LCL* HLA A2.1 heterozygous (A2.1, A29)  
Davies LCL* HLA A2.1 heterozygous (A1, A2.1)  
Hill Gaston LCL* HLA A2.1 negative (A1, Aw32)  
Kaur LCL* HLA A2.1 negative (A9)  
Leese LCL* HLA A2.1 negative (A3)  
Mutu I6154 Clone c59  
Mutu III61 Clone c62  
KM-H262  
HDLM-263  
L42864  
HS44565   
Cell Line Comments
B95.859 Marmoset cell line 
X50-760 Cord blood LCL  
Coneron LCL* HLA A2.1 homozygous  
Bevan LCL* HLA A2.1 homozygous  
M Rowe LCL* HLA A2.1 heterozygous (A2.1, A29)  
Davies LCL* HLA A2.1 heterozygous (A1, A2.1)  
Hill Gaston LCL* HLA A2.1 negative (A1, Aw32)  
Kaur LCL* HLA A2.1 negative (A9)  
Leese LCL* HLA A2.1 negative (A3)  
Mutu I6154 Clone c59  
Mutu III61 Clone c62  
KM-H262  
HDLM-263  
L42864  
HS44565   

*LCLs prepared in CRC laboratories, University of Birmingham, UK.

The demonstration of some antigens in paraffin material required microwave pretreatment in 0.1 mol/L citrate buffer, pH 6.0. The optimum microwave pretreatment times were determined for each antigen and are listed in Table 2. After pretreatment, endogenous peroxidase activity was blocked in 0.3% hydrogen peroxide in methanol and sections transferred to TBS.

Table 2.

Primary Antibodies Used in the Immunohistochemical Analyses, Together With Optimum Dilutions and Microwave Pretreatment Times

Antigen Antibody Species and Clone SourceOptimum Dilution Microwave Time (min)
LMP1 Mouse, CS1-4  Young et al,66 Dako Ltd. Cat No. M0897 1:500  20  
LMP2  Rat, 14B7-1-1  Gift from E. Kremmer, Munich, Germany22   
TAP-1  Mouse, Ring4  Gift from Dr J. Trowsdale, Oxford  1:1,000  0  
TAP-2  Mouse, AK-7  Gift from Dr J. Trowsdale, Oxford  1:500  0  
HLA A2.1 Mouse, MA2.1  Gift from Dr S. Lee, University of Birmingham 1:50  0  
HLA I  Mouse, HC-10  Gift from Dr P. Stern 1:50  20 
Antigen Antibody Species and Clone SourceOptimum Dilution Microwave Time (min)
LMP1 Mouse, CS1-4  Young et al,66 Dako Ltd. Cat No. M0897 1:500  20  
LMP2  Rat, 14B7-1-1  Gift from E. Kremmer, Munich, Germany22   
TAP-1  Mouse, Ring4  Gift from Dr J. Trowsdale, Oxford  1:1,000  0  
TAP-2  Mouse, AK-7  Gift from Dr J. Trowsdale, Oxford  1:500  0  
HLA A2.1 Mouse, MA2.1  Gift from Dr S. Lee, University of Birmingham 1:50  0  
HLA I  Mouse, HC-10  Gift from Dr P. Stern 1:50  20 

Sections were then incubated in primary antibodies at optimum dilutions previously determined for each antibody (Table 2). All primary antibodies were diluted in 10% normal sheep serum. Mouse monoclonal antibodies were detected either using the peroxidase-based Dako Duet system (Dako Ltd cat no: K492; Dako Ltd, Glostrup, Denmark) or the standard alkaline phosphatase and alkaline phosphatase (APAAP) method. The rat monoclonal antibody to LMP2 was detected using rabbit anti-rat immunoglobulins (Dako Ltd cat no: Z455) at a dilution of 1:400, followed by the Dako Catalyzed Signal Amplification system (Dako Ltd cat no:K0620).

Positive controls for LMP1 and LMP2 immunostaining consisted of paraffin-wax sections of LCL-SCID tumors. Positive controls for MHC class I, TAP-1, and TAP-2 included paraffin-wax and frozen sections of human tonsil. Negative controls for immunohistochemistry consisted of consecutive test sections in which the primary antibody was replaced with nonimmune serum of the same IgG subclass. Full details of antibodies used are given in Table 2.

Epitope sequencing.

A series of frozen HD biopsies were initially studied to determine their EBV and MHC status. The presence of latent EBV infection was determined as described above. HLA A2.1-positive tumor samples were identified in APAAP immunohistochemical assays using the MA2.1 antibody (Table 2). A variety of HLA A2.1-positive cell lines (both homozygous and heterozygous) and HLA A2.1-negative cell lines (Table 1) were used as controls in these immunohistochemical assays. Both acetone-fixed cytospin preparations and frozen sections of SCID tumors generated from these lines were tested.

HLA A2.1 and EBV-positive HD samples identified in this way were subjected to DNA extraction and PCR sequencing to determine the sequence of the LMP2A epitope, CLGGLLTMV, previously identified to be restricted through HLA A2.1. Sequences were compared with wild-type (B95.8) DNA. To obtain sufficient DNA from HD biopsies for sequencing it was necessary to extract DNA from agarose gels for reamplification. Primers, D5639LY and D5640LY (Fig 1A), were used in both PCR amplification and sequencing steps. Sequencing was performed twice in both directions on all samples.

Fig. 1.

(A) LMP2A sequence showing position of epitope and primers used in the PCR and sequencing analyses. (B) Autoradiograph of sequencing gel showing wild-type LMP2 sequence.

Fig. 1.

(A) LMP2A sequence showing position of epitope and primers used in the PCR and sequencing analyses. (B) Autoradiograph of sequencing gel showing wild-type LMP2 sequence.

Close modal
Detection of latent EBV gene expression.

EBER expression was shown in HRS cells of 18 of 39 examined HD cases. The expression of LMP1 in all EBER-positive HD cases was confirmed by immunohistochemistry. LMP2 protein was also detectable in the cytoplasm of HRS cells in 16 of 16 EBV-positive HD cases (Fig 2).

Fig. 2.

Immunohistochemical demonstration of LMP2 expression by HRS cells in a case of EBV-positive HD.

Fig. 2.

Immunohistochemical demonstration of LMP2 expression by HRS cells in a case of EBV-positive HD.

Close modal
MHC class I staining.

In all tissue sections, the presence of MHC class I-positive cells served as an internal control. Expression of MHC class I was mainly confined to the cell membrane of positively stained cells. Of 17 cases of EBER-positive HD analyzed, 13 showed the presence of MHC class I expression on HRS cells. However, MHC class I expression was detectable on HRS cells in only 4 of 21 cases of EBV-negative HD. In most EBV-positive HD samples, HRS cells could be easily identified because the intensity of class I staining was often stronger than on surrounding reactive cells (Fig 3A). This was in contrast to the majority of EBV-negative HD tumors, where HRS cells were either not stained or only weakly stained for class I (Fig3B). Class I expression was not related to subtype, and similar results were obtained on paraffin and frozen sections of HD tumors. In all cases, reactivity of surrounding reactive cells could be distinguished from staining on HRS cells. The HD cell lines, HDLM2, Km-H2, and HS445, all showed strong expression of MHC class I, whereas the HD cell line, L428, was consistently negative in all assays (not shown).

Fig. 3.

Immunohistochemistry for MHC class I expression. Long black arrows show class I expression on HRS cells (A) or its absence (B). The intensity of class I staining on surrounding small reactive cells (short red arrows) is less than that of the HRS cells in (A) and greater than that of the HRS cells in (B).

Fig. 3.

Immunohistochemistry for MHC class I expression. Long black arrows show class I expression on HRS cells (A) or its absence (B). The intensity of class I staining on surrounding small reactive cells (short red arrows) is less than that of the HRS cells in (A) and greater than that of the HRS cells in (B).

Close modal
Immunohistochemistry for TAP-1 and TAP-2.

On control tonsil tissue, germinal center cells were generally not stained for TAP-1 and TAP-2, with the exception of occasional positive cells. The interfollicular cells were most strongly stained for both markers and the squamous epithelium, when present, was also positively stained (data not shown). Similar results were obtained on both frozen and paraffin-wax sections. Cell preparations (both acetone-fixed cytospins and pelleted paraffin-wax sections) of Mutu I and Mutu III cell lines expressed both TAP-1 and TAP-2, although staining for both markers was noticeably weaker in Mutu I cells when compared with the Mutu III line. All the HD cell lines tested (HDLM2, L428, HS445, and Km-H2) were strongly stained for both TAP-1 and TAP-2. Frozen and paraffin wax-embedded HD tumors were also analyzed for TAP-1 and TAP-2 expression. With the exception of two HD cases, HRS cells showed strong staining for TAP-1 (Fig 4), irrespective of the presence of latent EBV infection in these cells. Selected HD cases were also stained for TAP-2. In all cases, HRS cells showed strong staining for TAP-2 (not shown).The results of EBER in situ hybridization, MHC I, and TAP-1 immunohistochemical assays are summarized in Table 3.

Fig. 4.

Immunohistochemistry for TAP-1 in HD by HRS cells.

Fig. 4.

Immunohistochemistry for TAP-1 in HD by HRS cells.

Close modal
Table 3.

Expression of MHC Class I and TAP-1 by HRS Cells in HD Specimens: Correlation With the Presence of EBV Within HRS Cells as Determined by EBER In Situ Hybridization (EBERs) and Subtype

Specimen No. Subtype EBER MHC Class I TAP-1
6828  MC  +  +  +  
2491/86  MC +  +  +  
116/80  MC  +  wk+  wk+  
2887/85 MC  +  +  +  
8040/81  MC  +  +  
444/85  MC  +  −  +  
976B5/86  MC  +  +  
3687-1/88  MC  +  +  +  
18331/84  MC  +  +  
E7926/89  MC  +  +  +  
5512/89  NS +  +  +  
1143/91  NS  +  +  +  
9535/90 NS  +  wk+  wk+  
9083/81  NS  +  nd  
963A/84  NS  +  +  +  
SAJ2891/89  NS  −  +  
1090/90S  NS  +  −  +  
1119B5/94S NS  +  −  +  
6696/87  LP  −  −  
2160/81  LP  −  −  +  
3513/80  MC  − ±  +  
83s/3610  MC  −  wk+  +  
2959S85 MC  −  −  +  
1633A/81  MC  −  −  
22300/83  MC  −  +  +  
9598/91  NS  − ±  +  
663/80  NS  −  −  −  
5165/83S NS  −  wk+  +  
7590-2/85  NS  −  −  
2153/87S  NS  −  −  +  
6870/87S  NS  − +  +  
1220/90  NS  −  −  +  
214F/90  NS −  −  +  
77/90  NS  −  −  
180B/93S  NS  −  −  −  
5758B/91  NS  − −  +  
388/91S  NS  −  −  +  
1307/84 NS  −  −  +  
Y1114B/89  NS  −  − 
Specimen No. Subtype EBER MHC Class I TAP-1
6828  MC  +  +  +  
2491/86  MC +  +  +  
116/80  MC  +  wk+  wk+  
2887/85 MC  +  +  +  
8040/81  MC  +  +  
444/85  MC  +  −  +  
976B5/86  MC  +  +  
3687-1/88  MC  +  +  +  
18331/84  MC  +  +  
E7926/89  MC  +  +  +  
5512/89  NS +  +  +  
1143/91  NS  +  +  +  
9535/90 NS  +  wk+  wk+  
9083/81  NS  +  nd  
963A/84  NS  +  +  +  
SAJ2891/89  NS  −  +  
1090/90S  NS  +  −  +  
1119B5/94S NS  +  −  +  
6696/87  LP  −  −  
2160/81  LP  −  −  +  
3513/80  MC  − ±  +  
83s/3610  MC  −  wk+  +  
2959S85 MC  −  −  +  
1633A/81  MC  −  −  
22300/83  MC  −  +  +  
9598/91  NS  − ±  +  
663/80  NS  −  −  −  
5165/83S NS  −  wk+  +  
7590-2/85  NS  −  −  
2153/87S  NS  −  −  +  
6870/87S  NS  − +  +  
1220/90  NS  −  −  +  
214F/90  NS −  −  +  
77/90  NS  −  −  
180B/93S  NS  −  −  −  
5758B/91  NS  − −  +  
388/91S  NS  −  −  +  
1307/84 NS  −  −  +  
Y1114B/89  NS  −  − 

Abbreviations: MC, mixed cellularity, NS, nodular sclerosis; LP, lymphocyte predominant, wk+, staining of HRS cells was weak only; ±, staining of HRS cells was equivocal; nd, not done.

Epitope sequencing.

A series of LCLs prepared from HLA A2.1-positive and -negative donors (Table 1) initially served as controls for the MA2.1 antibody. All cell lines from A2.1-positive donors reacted strongly with this antibody in APAAP assays, and all A2.1-negative cell lines, as expected, were nonreactive (data not shown).

Of the 20 HD biopsies available as frozen material, 9 contained EBER-positive HRS cells, and a further 6 of these were HLA A2.1 positive by immunohistochemistry. These 6 biopsies were subjected to PCR analysis to determine the sequence of the A2.1-restricted LMP2A-derived CTL epitope, CLGGLLMTV (Fig 1B). All 6 HD tumors showed wild-type epitope sequence.

There are strong indications that the outgrowth of EBV-infected tumors in vivo is dependent on the pattern of viral antigens expressed by the tumor and also the level of immunocompetence of the host. For example, the role of immunosurveillance in limiting the proliferation of EBV-infected cells in vivo is clearly illustrated by the development of EBV-positive lymphoproliferative disease in immunocompromised individuals.38-41 These lymphoproliferations may be regarded as the in vivo counterpart of LCLs. Indeed, these lesions often regress when immunosuppressive therapy is withdrawn or reduced.42,43 Many of these lesions express the full spectrum of EBV latent genes, with high cell surface expression of ICAM1 and LFA3 providing a wide range of potential targets for the immune system.44 It generally has been assumed that the sensitivity of these lesions to EBV-specific CTLs may be similar to LCLs. Evidence supporting this assumption has recently been provided by adoptive transfer experiments in which donor lymphocytes were used as a source of virus-specific CTLs to treat EBV lymphoproliferative disease in bone marrow transplant recipients.45,46 

In contrast to posttransplant lymphoproliferative disease and related conditions, HD arises in relatively immunocompetent hosts. The consistent expression of LMP1 and LMP2 in EBV-positive HD3,12,20,21 and the demonstration of effective CTL responses to epitopes derived from these proteins in vitro36,47,48 implies that defects in the antigen processing/recognition machinery may be a feature of these tumors. The mechanisms accounting for the failure of CTLs to eliminate EBV-infected HRS cells have not been established yet, although MHC class I downregulation by HRS cells, as originally demonstrated by Poppema and Visser,49 has been suggested as a possible mechanism.

This study has shown high-level expression of MHC class I molecules on the surface of HRS cells in EBV-associated HD cases, and much lower levels of expression or the absence of expression on HRS cells in EBV-negative samples, and is in accordance with the results of a previous study.50 These results suggest that MHC class I downregulation is unlikely to account for the failure of CTLs to eliminate the tumor cells, at least in EBV-positive cases. The mechanism responsible for downregulation or absence of MHC molecules in EBV-negative HD has yet to be determined. Unlike cervical carcinomas, where downregulation of MHC class I was associated with low-level expression of TAP molecules,32 downregulation of TAP molecules was not shown in this present study. The high-level expression of MHC class I molecules on EBV-positive HRS cells potentially could be mediated by LMP1 expression. LMP1 has been shown to upregulate class I expression when transfected into EBV-negative B-cell lines51 and epithelial cells (Dawson C. and L.S.Y., unpublished observations). LMP1 also has been shown to increase the stimulatory capacity of EBV-negative BL lines in allogeneic mixed lymphocyte cultures,51 to upregulate the expression of adhesion molecules,52 and to enhance the presentation of endogenous and exogenous antigens.53,54 

Another possible mechanism of CTL escape is suggested by the finding that memory CTL responses specific for variant epitopes in two HLA A11-restricted CTL epitopes (residues 399-408 and 416-424) from the EBNA3B protein were not detectable in HLA A11-positive Chinese donors infected with strain variants.34,35 This present study has established that a recently identified epitope in LMP2 that is restricted through HLA A2.155 is wild type in a small series of HLA A2-positive HD tumors, suggesting that mutation in virus epitope sequences may not be a common feature of EBV-positive HD, at least in this group of patients. Similar data showing that this same epitope is not commonly mutated has been presented by Bryden et al.56 The lack of mutation may have important implications for potential CTL therapy for HD patients, particularly because the CTL response to this epitope previously has been shown to be reactive against a range of virus isolates, including type-1 and type-2 Caucasian, Southeast Asian, New Guinean, and African isolates. Some of these isolates contained amino acid substitutions within the epitope, but were nevertheless recognized by CTLs raised against the B95.8 virus.36 The finding of similar, functionally conserved epitopes restricted through relatively common HLA alleles may be important for the future development of CTL therapy for EBV-associated HD.

Recent studies have shown that the HD cell line, HDLM2, is able to process and present epitopes from LMP1 and LMP2 in the context of multiple class I alleles including HLA A2 and is sensitive to lysis by EBV-specific CTLs.48 Furthermore, using autologous fibroblasts infected with a vaccinia recombinant encoding LMP2 as a target, the same authors were able to identify and expand LMP2-specific CTLs from the peripheral blood of an HD patient. However, recent data have pointed to the possibility of local suppression of EBV-specific immunity in the vicinity of the tumor that may confound certain immunotherapeutic strategies in HD. This was first suggested by a comparative study of EBV-specific responses in patients with EBV-positive and EBV-negative HD which failed to show virus-specific cytotoxicity in the tumor-infiltrating lymphocytes of six EBV-positive cases.57 However, CTL precursors were detected in the blood of one of the EBV-positive patients. A possible explanation of this finding was reported by Herbst et al,58 who showed that significantly higher proportions of EBV-positive HD cases contained interleukin-10 (IL-10)-expressing tumor cells when compared with EBV-negative tumors, suggesting that IL-10 production by HRS cells may be responsible for local downregulation of the CTL response in EBV-positive cases.58 

The results of this present study provide some encouragement for the pursuit of CTL therapy for EBV-associated HD. However, further work is required to establish if the microenvironment of EBV-positive HRS cells is responsible for the inhibition of virus-specific CTLs and whether this effect is likely to confound immunotherapeutic strategies targeted at EBV-positive HD patients.

Similar data regarding MHC class I and TAP expression in Hodgkin's disease have recently been presented.67 

R.F.A. is a Leukemia Society Scholar.

Supported by the Cancer Research Campaign and the National Institutes of Health Grants No. PO1 CA15396 and PO1 CA69266.

Address reprint requests to Richard F. Ambinder, MD, PhD, Johns Hopkins Oncology Center, 418 N Bond St, Baltimore, MD 21231.

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

1
Miller G: Epstein-Barr virus. Biology, pathogenesis, and medical aspects, in Fields BN, Knipe DM (eds): Virology (ed 2). New York, NY, Raven, 1990, p 1921
2
Tao
Q
Robertson
KD
Manns
A
Hildesheim
A
Ambinder
RF
Epstein-Barr virus (EBV) in endemic Burkitt's lymphoma: Molecular analysis of primary tumor tissue.
Blood
91
1998
1373
3
Niedobitek
G
Agathanggelou
A
Rowe
M
Jones
EL
Jones
DB
Turyaguma
P
Oryema
J
Wright
DH
Young
LS
Heterogeneous expression of Epstein-Barr virus latent proteins in endemic Burkitt's lymphoma.
Blood
86
1995
659
4
Wu
TC
Mann
RB
Epstein
J
MacMahon
E
Lee
WA
Charache
P
Hayward
SD
Kurman
RJ
Hayward
GS
Ambinder
RF
Abundant expression of EBER1 small nuclear RNA in nasopharyngeal carcinoma: A morphologically distinctive target for detection of Epstein-Barr virus in formalin-fixed paraffin-embedded carcinoma specimens.
Am J Pathol
138
1991
1461
5
Young
LS
Dawson
CW
Clark
D
Rupani
H
Busson
P
Tursz
T
Johnson
A
Rickinson
AB
Epstein-Barr virus gene expression in nasopharyngeal carcinoma.
J Gen Virol
69
1988
1051
6
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
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
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
9
Gledhill
S
Gallagher
A
Jones
DB
Krajewski
AS
Alexander
FE
Klee
E
Wright
DH
O'Brien
C
Onions
DE
Jarrett
RF
Viral involvement in Hodgkin's disease: Detection of clonal type A Epstein-Barr virus genomes in tumour samples.
Br J Cancer
64
1991
227
10
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
11
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
12
Pallesen
G
Hamilton-Dutoit
SJ
Rowe
M
Young
LS
Expression of Epstein-Barr virus (EBV) latent gene products in tumour cells of Hodgkin's disease.
Lancet
337
1991
320
13
Herbst
H
Dallenbach
F
Hummel
M
Niedobitek
G
Pileri
S
Muller-Lantzsch
N
Stein
H
Epstein-Barr virus latent membrane protein expression in Hodgkin and Reed-Sternberg cells.
Proc Natl Acad Sci USA
88
1991
4766
14
Gulley
ML
Eagan
PA
Quintanilla-Martinez
L
Picado
AL
Smir
BN
Childs
C
Dunn
CD
Craig
FE
Williams JW Jr
Banks
PM
Epstein-Barr virus DNA is abundant and monoclonal in the Reed-Sternberg cells of Hodgkin's disease: Association with mixed cellularity subtype and Hispanic American ethnicity.
Blood
83
1994
1595
15
Levine
PH
Ablashi
DV
Berard
CW
Carbone
PP
Waggoner
DE
Malan
L
Elevated antibody titers to Epstein-Barr virus in Hodgkin's disease.
Cancer
27
1971
416
16
Henderson
BE
Dworsky
R
Menck
H
Alena
B
Henle
W
Henle
G
Terasaki
P
Newell
GR
Rawlings
W
Kinnear
BK
Case-control study of Hodgkin's disease. II. Herpesvirus group antibody titers and HL-A type.
J Natl Cancer Inst
51
1973
1443
17
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
320
1989
689
18
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
19
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:1479, 1995 (suppl)
20
Murray
PG
Young
LS
Rowe
M
Crocker
J
Immunohistochemical diagnosis of the Epstein-Barr virus-encoded latent membrane protein in paraffin sections of Hodgkin's disease.
J Pathol
166
1992
1
21
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
22
Niedobitek
G
Kremmer
E
Herbst
H
Whitehead
L
Dawson
CW
Niedobitek
E
von Ostau
C
Rooney
N
Grasser
FA
Young
LS
Immunohistochemical detection of the Epstein-Barr virus-encoded latent membrane protein 2A in Hodgkin's disease and infectious mononucleosis.
Blood
90
1997
1664
23
Grasser
FA
Murray
PG
Kremmer
E
Klein
K
Remberger
K
Feiden
W
Reynolds
G
Niedobitek
G
Young
LS
Mueller-Lantzsch
N
Monoclonal antibodies directed against the Epstein-Barr virus-encoded nuclear antigen 1 (EBNA1): Imunohistologic detection of EBNA1 in the malignant cells of Hodgkin's disease.
Blood
84
1994
3792
24
Gavioli
R
Kurilla
MG
De Campos-Lima
PO
Wallace
LE
Dolcetti
R
Murray
RJ
Rickinson
AB
Masucci
MG
Multiple HLA A11-resricted cytotoxic T lymphocyte epitopes of different immunogenicities in the EBV-encoded nuclear antigen 4.
J Virol
67
1993
1572
25
Levitskaya
J
Coram
M
Levitsky
V
Imreh
S
Steigerwaldmullen
PM
Klein
G
Kurilla
MG
Masucci
MG
Inhibition of antigen processing by the internal repeat region of the Epstein-Barr virus nuclear antigen-1.
Nature
375
1995
685
26
Khanna
R
Burrows
SR
Kurilla
MG
Jacob
CA
Misko
IS
Sculley
TB
Kieff
E
Moss
DJ
Localization of Epstein-Barr virus cytotoxic T cell epitopes using recombinant vaccinia: Implications for vaccine development.
J Exp Med
176
1992
169
27
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
28
Imreh
MP
Zhang
QJ
De Campos-Lima
PO
Imreh
S
Krausa
P
Browning
M
Klein
G
Masucci
MG
Mechanisms of allele-selective down-regulation of HLA class I in Burkitt's lymphoma.
Int J Cancer
62
1995
90
29
Masucci
MG
Klein
E
Cell phenotype dependent expression of MHC class I antigens in Burkitt's lymphoma cell lines.
Semin Cancer Biol
2
1991
63
30
Cromme
FV
van Bommel
PF
Walboomers
JM
Gallee
MP
Stern
PL
Kenemans
P
Helmerhorst
TJ
Stukart
MJ
Meijer
CJ
Differences in MHC and TAP-1 expression in cervical cancer lymph node metastases as compared with the primary tumours.
Br J Cancer
69
1994
1176
31
Cromme
FV
Snijders
PJ
van den Brule
AJ
Kenemans
P
Meijer
CJ
Walboomers
JM
HC class I expression in HPV 16 positive cervical carcinomas is post-transcriptionally controlled and independent from c-myc overexpression.
Oncogene
8
1993
2969
32
Cromme
FV
Airey
J
Heemels
MT
Ploegh
HL
Keating
PJ
Stern
PL
Meijer
CJ
Walboomers
JM
Loss of transporter protein, encoded by the TAP-1 gene, is highly correlated with loss of HLA expression in cervical carcinomas.
J Exp Med
179
1994
335
33
Trowsdale
J
Hanson
I
Mockridge
I
Beck
S
Townsend
A
Kelly
A
Sequences encoded in the class II region of the MHC related to the ‘ABC’ superfamily of transporters.
Nature
348
1990
741
34
De Campos-Lima
PO
Gavioli
R
Zhang
QJ
Wallace
LE
Dolcetti
R
Rowe
M
Rickinson
AB
Masucci
MG
HLA-A11 epitope loss in isolates of Epstein-Barr virus from a highly A11+ population.
Science
260
1993
98
35
De Campos-Lima
PO
Levitsky
V
Brooks
J
Lee
SP
Hu
LF
Rickinson
AB
Masucci
MG
T cell responses and virus evolution: Loss of HLA A11-restricted CTL epitopes in Epstein-Barr virus isolates from highly A11-positive populations by selective mutation of anchor residues.
J Exp Med
179
1994
1297
36
Lee
SP
Tierney
RJ
Thomas
WA
Brooks
JM
Rickinson
AB
Conserved CTL epitopes within EBV latent membrane protein 2: A potential target for CTL-based tumor therapy.
J Immunol
158
1997
3325
37
Barletta
JM
Kingma
DW
Ling
Y
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
38
Nalesnik
MA
Jaffe
R
Starzl
TE
Demetris
AJ
Porter
K
Burnham
JA
Makowka
L
Ho
M
Locker
J
The pathology of posttransplant lymphoproliferative disorders occurring in the setting of cyclosporine A-prednisone immunosuppression.
Am J Pathol
133
1988
173
39
Knowles
DM
Cesarman
E
Chadburn
A
Frizzera
G
Chen
J
Rose
EA
Michler
RE
Correlative morphologic and molecular genetic analysis demonstrates three distinct categories of posttransplantation lymphoproliferative disorders.
Blood
85
1995
552
40
Filipovich AH, Mertens A, Robison L, Ambinder RF, Shapiro RS, Frizzera G: Lymphoproliferative disorders associated with primary immunofeficiencies, in Magrath I (ed): The Non-Hodgkin's Lymphomas. New York, NY, Oxford, 1995, p 459
41
Hamilton-Dutoit
SJ
Rea
D
Raphael
M
Sandvej
K
Delecluse
HJ
Gisselbrecht
C
Marelle
L
van Krieken
HJ
Pallesen
G
Epstein-Barr virus-latent gene expression and tumor cell phenotype in acquired immunodeficiency syndrome-related non-Hodgkin's lymphoma. Correlation of lymphoma phenotype with three distinct patterns of viral latency.
Am J Pathol
143
1993
1072
42
Starzl
TE
Porter
KA
Iwatsuki
S
Rosenthal
JT
Shaw BW Jr
Atchison
RW
Nalesnik
MA
Ho
M
Griffith
BP
Hakala
TR
Hardesty
RL
Jaffe
R
Bahnson
HT
Reversibility of lymphomas and lymphoproliferative lesions developing under cyclosporin-steroid therapy.
Lancet
1
1984
583
43
Crawford
DH
Thomas
JA
Janossy
G
Sweny
P
Fernando
ON
Moorhead
JF
Thompson
JH
Epstein Barr virus nuclear antigen positive lymphoma after cyclosporin A treatment in patient with renal allograft.
Lancet
1
1980
1355
44
Thomas
JA
Hotchin
NA
Allday
MJ
Amlot
P
Rose
M
Yacoub
M
Crawford
DH
Immunohistology of Epstein-Barr virus-associated antigens in B cell disorders from immunocompromised individuals.
Transplantation
49
1990
944
45
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'Reilly
RJ
Infusions of donor leukocytes to treat Epstein-Barr virus-associated lymphoproliferative disorders after allogeneic bone marrow transplantation.
N Engl J Med
33
1994
1185
46
Rooney
CM
Smith
CA
Ng
CY
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
47
Rickinson
AB
Moss
DJ
Human cytotoxic T lymphocyte responses to Epstein-Barr virus infection.
Annu Rev Immunol
15
1997
405
48
Sing
AP
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 of EBV+ Hodgkin's disease.
Blood
89
1997
1978
49
Poppema
S
Visser
L
Absence of HLA class I expression by Reed-Sternberg cells.
Am J Pathol
145
1994
37
50
Oudejans
JJ
Jiwa
NM
Kummer
JA
Horstman
A
Vos
W
Baak
JPA
Kluin
PM
Vandervalk
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 Hodgkins disease.
Blood
87
1996
3844
51
Cuomo
L
Trivedi
P
Wang
F
Wimberg
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
52
Wang
F
Gregory
C
Sample
C
Rowe
M
Liebowitz
D
Murray
R
Rickinson
A
Kieff
E
Epstein-Barr virus latent membrane protein (LMP1) and nuclear proteins 2 and 3C are effectors of phenotypic changes in B lymphocytes: EBNA-2 and LMP1 cooperatively induce CD23.
J Virol
64
1990
2309
53
Rowe
M
Khanna
R
Jacob
CA
Argaet
V
Kelly
A
Powis
S
Belich
M
Croom-Carter
D
Lee
S
Burrows
SR
Trowsdale
J
Moss
DJ
Rickinson
AB
Restoration of endogenous antigen processing in Burkitt's lymphoma cells by Epstein-Barr virus latent membrane protein-1: Coordinate up-regulation of peptide transporters and HLA-class I antigen expression.
Eur J Immunol
25
1995
1374
54
De Campos-Lima
PO
Torsteindottir
S
Cuomo
L
Klein
G
Sulitzeanu
D
Masucci
M
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
55
Lee
SP
Thomas
WA
Murray
RJ
Khanim
F
Kaur
S
Young
LS
Rowe
M
Kurilla
M
Rickinson
AB
HLA A2.1-restricted cytotoxic T cells recognizing a range of Epstein-Barr virus isolates through a defined epitope in latent membrane protein LMP2.
J Virol
67
1993
7428
56
Bryden
H
MacKenzie
J
Andrew
L
Alexander
FE
Angus
B
Krajewski
AS
Armstrong
AA
Gray
D
Cartwright
RA
Kane
E
Wright
DH
Taylor
P
Jarrett
RF
Determination of HLA-A*02 antigen status in Hodgkin's disease and analysis of an HLA-A*02-restricted epitope of the Epstein-Barr virus LMP-2 protein.
Int J Cancer
72
1997
614
57
Frisan
T
Sjoberg
J
Dolcetti
R
Boiocchi
M
De Re
V
Carbone
A
Brautbar
C
Battat
S
Biberfeld
P
Eckman
M
Ost
O
Christensson
B
Sundstrom
C
Bjorkholm
M
Pisa
P
Masucci
MG
Local suppression of Epstein-Barr virus (EBV)-specific cytotoxicity in biopsies of EBV-positive Hodgkin's disease.
Blood
86
1995
1493
58
Herbst
H
Foss
HD
Samol
J
Araujo
I
Klotzbach
H
Krause
H
Agathanggelou
A
Niedobitek
G
Stein
H
Frequent expression of interleukin-10 by Epstein-Barr virus-harboring tumor cells of Hodgkin's disease.
Blood
87
1996
2918
59
Miller
G
Shope
T
Lisco
H
Stitt
D
Lipman
M
Epstein-Barr virus: Transformation, cytopathic changes, and viral antigens in squirrel monkey and marmoset leukocytes.
Proc Natl Acad Sci USA
69
1972
383
60
Yandava
CN
Speck
SH
Characterization of the deletion and rearrangement in the BamHI C region of the X50-7 Epstein-Barr virus genome, a mutant viral strain which exhibits constitutive BamHI W promoter activity.
J Virol
66
1992
5646
61
Gregory
CD
Rowe
M
Rickinson
AB
Different Epstein-Barr virus-B cell interactions in phenotypically distinct clones of a Burkitt's lymphoma cell line.
J Gen Virol
71
1990
1481
62
Kamesaki
H
Fukuhara
S
Tatsumi
E
Uchino
H
Yamabe
H
Miwa
H
Shirakawa
S
Hatanaka
M
Honjo
T
Cytochemical, immunologic, chromosomal, and molecular genetic analysis of a novel cell line derived from Hodgkin's disease.
Blood
68
1986
285
63
Drexler
HG
Gaedicke
G
Lok
MS
Diehl
V
Minowada
J
Hodgkin's disease derived cell lines HDLM-2 and L-428: Comparision of morphology, immunological and isoenzyme profiles.
Leuk Res
10
1986
487
64
Schaadt
M
Diehl
V
Stein
H
Fonatsch
C
Kirchner
HH
Two neoplastic cell lines with unique features derived from Hodgkin's disease.
Int J Cancer
26
1980
723
65
Klein
S
Jucker
M
Diehl
V
Tesch
H
Production of multiple cytokines by Hodgkin's disease derived cell lines.
Hematol Oncol
10
1992
319
66
Young
L
Alfieri
C
Hennessy
K
Evans
H
O'Hara
C
Anderson
KC
Ritz
J
Shapiro
RS
Rickinson
A
Kieff
E
Cohen
JI
Expression of Epstein-Barr virus transformation-associated genes in tissues of patients with EBV lymphoproliferative disease.
N Engl J Med
321
1989
1080
67
Lee
SP
Constandinou
CM
Thomas
WA
Croom-Carter
D
Blake
NW
Murray
PG
Crocker
J
Rickinson
AB
Antigen presenting phenotype of Hodgkin Reed-Sternberg cells: Analysis of the HLA class I processing pathway and the effects of interleukin-10 on Epstein-Barr virus-specific cytotoxic T-cell recognition.
Blood
92
1998
1020
Sign in via your Institution