Although a pathogenic role for the Epstein-Barr virus (EBV) is largely undisputed for tumors that are consistently EBV genome positive (eg, nasopharyngeal carcinoma, endemic Burkitt lymphoma), this is not the case for classical Hodgkin lymphoma (cHL), a tumor with only a variable EBV association. In light of recent developments in immunotherapeutics and small molecules targeting EBV, we believe it is now timely to reevaluate the role of EBV in cHL pathogenesis.

Classical Hodgkin lymphoma (cHL) is characterized by rare malignant Hodgkin/Reed Sternberg (HRS) cells in an inflammatory tumor microenvironment (TME). HRS cells are B cells with evidence of somatic hypermutation indicating that they are germinal center (GC) experienced.1  However, phenotypically, HRS cells do not resemble B cells and lack a functional B cell receptor (BCR); in ∼25% of cases, this is caused by mutations that destroy the coding capacity of the immunoglobulin genes.2 

The detection of raised antibody levels to EBV antigens in cHL patients provided the first evidence that EBV might be involved in cHL.3  These raised levels precede cHL development,4  an effect specific to virally associated cases (Figure 1).5  People with a prior history of infectious mononucleosis (IM) also have an increased cHL risk6,7  that is specific to EBV+ cases.8,9  EBV+ cHL is temporally related to primary infection, developing a median of 2.9 years after IM.8  In children, EBV+ cHL, but not EBV cHL, is associated with a seasonal peak in the onset of symptoms, consistent with an increased risk for the tumor following primary infection.10 

Figure 1.

Risk factors associated with the development of EBV+ cHL.

Figure 1.

Risk factors associated with the development of EBV+ cHL.

Close modal

An increased and decreased risk of EBV+ cHL is also associated with HLA-A*01 and HLA-A*02 alleles, respectively.11-15  Although the same genotypic markers that confer a risk for EBV+ cHL are associated with IM risk, HLA-A typing has revealed that a prior history of IM is independently associated with EBV+ cHL, even after adjusting for the effects of HLA-A alleles.

The risk factors associated with cHL have been identified almost exclusively in individuals from resource-rich nations. However, we do not know whether these risk factors are the same for cHL occurring in resource-poor countries (eg, in sub-Saharan Africa where IM is rare [>80% of children are infected at younger than 5 years of age]16  but 70% to 90% of cHL is EBV+).17,18 

Infection of the tumor cells is considered a sine qua non for establishing a causal association between virus and tumor. In cHL, these conditions were fulfilled shortly after the initial serological analyses had implicated EBV. Thus, in 1985, the EBNA protein (Epstein-Barr nuclear antigen, now known to be EBNA1) was detected in HRS cells.19  EBV DNA was detected in 20% to 25% of Hodgkin lymphoma biopsies by Southern blotting,20  and in situ hybridization for EBV DNA confirmed the EBV genome in HRS cells.21,22  Later, the abundant untranslated Epstein-Barr encoded RNAs (EBER1 and EBER2) would provide a target for the routine detection of EBV in tumors, including cHL.23  Finally, HRS cells were shown to express the EBV-encoded latent membrane protein-1 (LMP1), LMP2A, and EBNA1 proteins.24-27 

Viral genomes are monoclonal in cHL. Upon cell entry, the linear EBV DNA circularizes by recombination of the terminal repeats (TRs), producing fused termini of a unique length for each circularization event. Detection of multiple TR lengths within a sample specifies a polyclonal infection, whereas TRs with an identical number of repeats indicate expansion of a single infected progenitor cell. Thus, the detection of a single TR length in cHL strongly argues for an etiological role for EBV.21  Moreover, the number of EBV genomes present per cell is different between patients but is relatively constant in different samples from the same patient.28  EBV also persists in HRS cells throughout the course of disease and is present in multiple sites of disease28  (Figure 2). These are important observations; EBV episomes can be lost by asymmetrical segregation during cell divisions with retention of EBV genomes in cHL suggesting that the virus provides an ongoing advantage to the tumor cells.

Figure 2.

Molecular evidence of a pathogenic role for EBV in cHL.

Figure 2.

Molecular evidence of a pathogenic role for EBV in cHL.

Close modal

The frequent detection of EBV in cHL and, in these cases, the presence of the virus in every tumor cell is strong evidence of an etiological association. EBV is present in a third of cHL cases in the west; the EBV association is even higher in some parts of the world. Were the virus simply a passenger, then the frequency of EBV+ cHL would approximate the number of EBV-infected B cells in an individual (ie, ∼1 in 106 of the total B cell pool). If we repeat this calculation, this time assuming that EBV+ cHL arises only from GC B cells, the expected frequency of EBV+ cHL would be equivalent to the frequency of EBV-infected B cells in the GC (ie, 4.6 per 104, using the highest reported frequency of EBV infection in GC B cells).29 

The geographical patterns of cancer incidence can indicate an infectious etiology. Resource-rich nations generally have a lower burden of virally related cancers than resource-limited countries.30,31  Thus, EBV rates in cHL from North America and Europe are lower than in resource-limited countries.32,33  EBV+ rates for cHL are also higher in Asians and Hispanics compared with whites or blacks,30  as well as in South Asian children compared with non-South Asian children in the UK.34  In resource-rich countries, the proportion of cases with EBV is highest in older people and children, with lower rates in young adults.35,36  The global patterns of EBV+ cHL differ somewhat from those observed for EBV+ Burkitt lymphoma which is characterized by very high rates in so-called “endemic” areas but much lower rates across the rest of world.

Virus-associated cancers are increased in immunosuppressed persons (exemplified by posttransplant lymphoproliferative disease; PTLD) and in older people, an effect attributed to an age-related decline in virus-specific immunity.35  However, EBV+ cHL is the least common major form of PTLD. Moreover, although the incidence of EBV+ cHL is increased in HIV infection,37,38  this is only observed in patients with intermediate levels of immune impairment.37  Moreover, the incidence of cHL in HIV+ patients has not fallen in the era of highly active antiretroviral therapy; indeed, cHL risk may be increased following immune reconstitution on highly active antiretroviral therapy.39-41  This apparent paradox can be explained by the dependency of HRS cells on CD4+ T cells.

Three EBV-encoded proteins and viral microRNA (miRNA) expressed in virus-infected HRS cells are implicated in cHL pathogenesis.

EBNA1 is essential to maintain EBV infection, acting as a viral replication factor and tether between the viral genome and the host cell chromosomes ensuring (for the most part) faithful segregation of viral episomes during cell division.42  EBNA1 is also a transcription factor that regulates viral and cellular gene expression.43-45  EBNA1 can promote the growth and survival of HRS cells, an effect attributable to the downregulation of the protein tyrosine phosphatase receptor type K.46,47 

LMP1 is a constitutively active CD40 receptor homolog48  that induces many of the gene expression changes characteristic of HRS cells.49-51  LMP1 induces multiple cell signaling pathways that are aberrantly activated in HRS cells, including NF-κB and JAK/STAT.52-55  Alternatively, these pathways can be activated by cellular mutations in cHL.56  Accordingly, EBV+ cHL has significantly fewer cellular mutations,57  including mutations in negative regulators of the NF-κB pathway (eg, TNFAIP3, NFKBIA).58-60  EBV+ tumors also have fewer chromosomal breakpoints and aneuploid autosomes.61  Thus, in EBV+ cHL, viral genes are thought to be sufficient to drive aberrant activation of signaling pathways in the absence of cellular mutations, indicating that EBV and cellular mutations provide mutually exclusive means to the same pathogenic end point.

A variant of LMP1, carrying a 30–base pair deletion in the C terminus, reported to have enhanced transforming properties62,63  has been found more commonly in HIV+ cHL compared with non-HIV–associated cHL,64  as well as in pediatric cHL compared with normal controls.65  A study of HIV patients also identified LMP1 polymorphisms associated with increased NF-κB activation, but these variants were not found to be significantly enriched among EBV+ cHL.66 

LMP2A is a BCR mimic that allows B-cell development in the absence of normal BCR signaling.67,68  EBV can immortalize BCR GC B cells, an effect that is dependent upon LMP2A.69-72  Consistent with this, crippling mutations in immunoglobulin genes are found almost exclusively in EBV+ cHL.73  LMP2A also suppresses expression of B-cell transcription factors, including EBF1 and E2A, in turn recapitulating many of the gene expression changes seen in HRS cells.74-77 

The EBV latent genes probably also contribute to the characteristic TME of cHL. Thus, LMP1 can recruit cells of the TME by inducing the expression of numerous chemokines and cytokines in infected HRS cells.78-80  Similarly, EBNA1 has been shown to increase expression of the T-cell chemokine CCL20 and, in turn, the recruitment of regulatory T cells.81 

Loss of HLA expression is common in HRS cells, yet, counterintuitively, HLA class I loss is more frequent in EBV cHL,82-84  which, in nodular sclerosis cases, could be due to mutations in the β-2 microglobulin gene.85  In contrast, EBV+ cHL expresses normal or even elevated levels of HLA and contains more activated cytotoxic T lymphocytes than EBV cHL.82-84,86  Other mechanisms could explain immune evasion in EBV+ cHL. For example, LMP1 has been shown to induce PD-L1 expression to suppress cytotoxic T cell responses.87 

EBV-encoded BART miRNAs are also expressed in EBV+ cHL88  and could contribute to lymphomagenesis by regulating viral and cellular gene expression; BART miRNAs have effects on innate immunity (eg, miR-BART6-3p targets DDX58 encoding retinoic acid–inducible protein 1, a sensor of viral messenger RNA),89  adaptive immunity (eg, miR-BART1 and miR-BART2, which reduce antigen presentation, and miR-BART17, which targets TAP2),90,91  apoptosis (eg, multiple BART miRNAs targeting caspase 3),92,93  and proliferation (eg, miR-BART6-3p targets PTEN).94 

The detection of EBV within cHL, although sometimes useful in confirming the diagnosis, does not influence patient management. Combination chemotherapy and radiotherapy is the mainstay of treatment of cHL patients, with 5-year survival rates in the range of 80% to 90%. However, the prognosis for patients with refractory disease or for those who relapse after conventional therapy is dismal. The current treatments also cause significant long-term toxicities that include secondary malignancies, cardiopulmonary toxicity, hypothyroidism, and infertility. Outcomes are particularly poor for older patients (5-year survival 30-50% in patients older than 70 years).95  Furthermore, an EBV+ status in elderly patients is associated with an even worse prognosis.96-100 

It is in this context that immune checkpoint inhibitors targeting the PD-1 pathway are providing impressive clinical results, even in heavily pretreated patients.87,101-104  Further improvements in these clinical responses could be achieved by combining immune checkpoint inhibitors with EBV-specific therapies. Ex vivo–manufactured donor T cells and patient-derived EBV-specific T cells are already available and have proven effective, particularly in patients with PTLD.105,106  More recently, vaccine approaches have been developed based on recombinant virus proteins or virus peptides.107,108  Furthermore, recent developments in the identification and design of EBNA1 inhibitors have opened up new possibilities to therapeutically target EBV-associated malignancies, including cHL.109-112 

Although some cHL patients harbor EBV in tumor cells, the contribution of the virus to this tumor has been the subject of debate. Differences in the epidemiology, genetics, biology, and clinical behavior of EBV+ vs EBV cHL strongly indicate a pathogenic role for the virus. A detailed understanding of the relationship between EBV and cHL provides insights into cHL pathogenesis and highlights opportunities for novel therapeutic interventions.

This work was supported by Bloodwise.

Contribution: P.G.M. and L.S.Y. conceived the idea and wrote the manuscript.

Conflict-of-interest disclosure: L.S.Y. is a member of the Scientific Advisory Board of Viracta Therapeutics, Inc. P.G.M. declares no competing financial interests.

Correspondence: Paul G. Murray, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Dr, Birmingham B15 2TT, United Kingdom; and Health Research Institute, University of Limerick, Limerick V94 T9PX, Ireland; e-mail: p.g.murray@bham.ac.uk.

1.
Küppers
R
,
Rajewsky
K
,
Zhao
M
, et al
.
Hodgkin disease: Hodgkin and Reed-Sternberg cells picked from histological sections show clonal immunoglobulin gene rearrangements and appear to be derived from B cells at various stages of development
.
Proc Natl Acad Sci USA
.
1994
;
91
(
23
):
10962
-
10966
.
2.
Kanzler
H
,
Küppers
R
,
Hansmann
ML
,
Rajewsky
K
.
Hodgkin and Reed-Sternberg cells in Hodgkin’s disease represent the outgrowth of a dominant tumor clone derived from (crippled) germinal center B cells
.
J Exp Med
.
1996
;
184
(
4
):
1495
-
1505
.
3.
Levine
PHAD
,
Ablashi
DV
,
Berard
CW
,
Carbone
PP
,
Waggoner
DE
,
Malan
L
.
Elevated antibody titers to Epstein-Barr virus in Hodgkin’s disease
.
Cancer
.
1971
;
27
(
2
):
416
-
421
.
4.
Mueller
N
,
Evans
A
,
Harris
NL
, et al
.
Hodgkin’s disease and Epstein-Barr virus. Altered antibody pattern before diagnosis
.
N Engl J Med
.
1989
;
320
(
11
):
689
-
695
.
5.
Levin
LI
,
Chang
ET
,
Ambinder
RF
, et al
.
Atypical prediagnosis Epstein-Barr virus serology restricted to EBV-positive Hodgkin lymphoma
.
Blood
.
2012
;
120
(
18
):
3750
-
3755
.
6.
Connelly
RR
,
Christine
BW
.
A cohort study of cancer following infectious mononucleosis
.
Cancer Res
.
1974
;
34
(
5
):
1172
-
1178
.
7.
Rosdahl
N
,
Larsen
SO
,
Clemmesen
J
.
Hodgkin’s disease in patients with previous infectious mononucleosis: 30 years’ experience
.
BMJ
.
1974
;
2
(
5913
):
253
-
256
.
8.
Hjalgrim
H
,
Smedby
KE
,
Rostgaard
K
, et al
.
Infectious mononucleosis, childhood social environment, and risk of Hodgkin lymphoma
.
Cancer Res
.
2007
;
67
(
5
):
2382
-
2388
.
9.
Hjalgrim
H
,
Munksgaard
L
,
Melbye
M
.
[Epstein-Barr virus and Hodgkin’s lymphoma]
.
Ugeskr Laeger
.
2002
;
164
(
50
):
5924
-
5927
.
10.
Reiman
A
,
Powell
JE
,
Flavell
KJ
, et al
.
Seasonal differences in the onset of the EBV-positive and -negative forms of paediatric Hodgkin’s lymphoma
.
Br J Cancer
.
2003
;
89
(
7
):
1200
-
1201
.
11.
Kushekhar
K
,
van den Berg
A
,
Nolte
I
,
Hepkema
B
,
Visser
L
,
Diepstra
A
.
Genetic associations in classical Hodgkin lymphoma: a systematic review and insights into susceptibility mechanisms
.
Cancer Epidemiol Biomarkers Prev
.
2014
;
23
(
12
):
2737
-
2747
.
12.
Hjalgrim
H
,
Rostgaard
K
,
Johnson
PC
, et al
.
HLA-A alleles and infectious mononucleosis suggest a critical role for cytotoxic T-cell response in EBV-related Hodgkin lymphoma
.
Proc Natl Acad Sci USA
.
2010
;
107
(
14
):
6400
-
6405
.
13.
Cozen
W
,
Timofeeva
MN
,
Li
D
, et al
.
A meta-analysis of Hodgkin lymphoma reveals 19p13.3 TCF3 as a novel susceptibility locus
.
Nat Commun
.
2014
;
5
(
1
):
3856
.
14.
Niens
M
,
Jarrett
RF
,
Hepkema
B
, et al
.
HLA-A*02 is associated with a reduced risk and HLA-A*01 with an increased risk of developing EBV+ Hodgkin lymphoma
.
Blood
.
2007
;
110
(
9
):
3310
-
3315
.
15.
Niens
M
,
van den Berg
A
,
Diepstra
A
, et al
.
The human leukocyte antigen class I region is associated with EBV-positive Hodgkin’s lymphoma: HLA-A and HLA complex group 9 are putative candidate genes
.
Cancer Epidemiol Biomarkers Prev
.
2006
;
15
(
11
):
2280
-
2284
.
16.
Piriou
E
,
Asito
AS
,
Sumba
PO
, et al
.
Early age at time of primary Epstein-Barr virus infection results in poorly controlled viral infection in infants from Western Kenya: clues to the etiology of endemic Burkitt lymphoma
.
J Infect Dis
.
2012
;
205
(
6
):
906
-
913
.
17.
Lee
JH
,
Kim
Y
,
Choi
JW
,
Kim
YS
.
Prevalence and prognostic significance of Epstein-Barr virus infection in classical Hodgkin’s lymphoma: a meta-analysis
.
Arch Med Res
.
2014
;
45
(
5
):
417
-
431
.
18.
Stefan
DC
.
Hodgkin lymphoma in Africa: present and future
.
Transfus Apheresis Sci
.
2013
;
49
(
2
):
144
-
146
.
19.
Poppema
S
,
van Imhoff
G
,
Torensma
R
,
Smit
J
.
Lymphadenopathy morphologically consistent with Hodgkin’s disease associated with Epstein-Barr virus infection
.
Am J Clin Pathol
.
1985
;
84
(
3
):
385
-
390
.
20.
Weiss
LM
,
Strickler
JG
,
Warnke
RA
,
Purtilo
DT
,
Sklar
J
.
Epstein-Barr viral DNA in tissues of Hodgkin’s disease
.
Am J Pathol
.
1987
;
129
(
1
):
86
-
91
.
21.
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
.
1989
;
74
(
2
):
810
-
816
.
22.
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
.
1989
;
320
(
8
):
502
-
506
.
23.
Wu
T-C
,
Mann
RB
,
Charache
P
, et al
.
Detection of EBV gene expression in Reed-Sternberg cells of Hodgkin’s disease
.
Int J Cancer
.
1990
;
46
(
5
):
801
-
804
.
24.
Grässer
FA
,
Murray
PG
,
Kremmer
E
, et al
.
Monoclonal antibodies directed against the Epstein-Barr virus-encoded nuclear antigen 1 (EBNA1): immunohistologic detection of EBNA1 in the malignant cells of Hodgkin’s disease
.
Blood
.
1994
;
84
(
11
):
3792
-
3798
.
25.
Pallesen
G
,
Hamilton-Dutoit
SJ
,
Rowe
M
,
Young
LS
.
Expression of Epstein-Barr virus latent gene products in tumour cells of Hodgkin’s disease
.
Lancet
.
1991
;
337
(
8737
):
320
-
322
.
26.
Deacon
EM
,
Pallesen
G
,
Niedobitek
G
, et al
.
Epstein-Barr virus and Hodgkin’s disease: transcriptional analysis of virus latency in the malignant cells
.
J Exp Med
.
1993
;
177
(
2
):
339
-
349
.
27.
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
.
1992
;
166
(
1
):
1
-
5
.
28.
Coates
PJ
,
Slavin
G
,
D’Ardenne
AJ
.
Persistence of Epstein-Barr virus in Reed-Sternberg cells throughout the course of Hodgkin’s disease
.
J Pathol
.
1991
;
164
(
4
):
291
-
297
.
29.
Roughan
JE
,
Torgbor
C
,
Thorley-Lawson
DA
.
Germinal center B cells latently infected with Epstein-Barr virus proliferate extensively but do not increase in number
.
J Virol
.
2010
;
84
(
2
):
1158
-
1168
.
30.
Glaser
SL
,
Lin
RJ
,
Stewart
SL
, et al
.
Epstein-Barr virus-associated Hodgkin’s disease: epidemiologic characteristics in international data
.
Int J Cancer
.
1997
;
70
(
4
):
375
-
382
.
31.
Glaser
SL
,
Jarrett
RF
.
The epidemiology of Hodgkin’s disease
.
Baillieres Clin Haematol
.
1996
;
9
(
3
):
401
-
416
.
32.
Chang
KL
,
Albújar
PF
,
Chen
YY
,
Johnson
RM
,
Weiss
LM
.
High prevalence of Epstein-Barr virus in the Reed-Sternberg cells of Hodgkin’s disease occurring in Peru
.
Blood
.
1993
;
81
(
2
):
496
-
501
.
33.
Weinreb
M
,
Day
PJ
,
Niggli
F
, et al
.
The consistent association between Epstein-Barr virus and Hodgkin’s disease in children in Kenya
.
Blood
.
1996
;
87
(
9
):
3828
-
3836
.
34.
Flavell
KJ
,
Biddulph
JP
,
Powell
JE
, et al
.
South Asian ethnicity and material deprivation increase the risk of Epstein-Barr virus infection in childhood Hodgkin’s disease
.
Br J Cancer
.
2001
;
85
(
3
):
350
-
356
.
35.
Armstrong
AA
,
Alexander
FE
,
Cartwright
R
, et al
.
Epstein-Barr virus and Hodgkin’s disease: further evidence for the three disease hypothesis
.
Leukemia
.
1998
;
12
(
8
):
1272
-
1276
.
36.
Jarrett
RF
,
Gallagher
A
,
Jones
DB
, et al
.
Detection of Epstein-Barr virus genomes in Hodgkin’s disease: relation to age
.
J Clin Pathol
.
1991
;
44
(
10
):
844
-
848
.
37.
Biggar
RJ
,
Jaffe
ES
,
Goedert
JJ
,
Chaturvedi
A
,
Pfeiffer
R
,
Engels
EA
.
Hodgkin lymphoma and immunodeficiency in persons with HIV/AIDS
.
Blood
.
2006
;
108
(
12
):
3786
-
3791
.
38.
Glaser
SL
,
Clarke
CA
,
Gulley
ML
, et al
.
Population-based patterns of human immunodeficiency virus-related Hodgkin lymphoma in the Greater San Francisco Bay Area, 1988-1998
.
Cancer
.
2003
;
98
(
2
):
300
-
309
.
39.
Bohlius
J
,
Schmidlin
K
,
Boué
F
, et al
; Collaboration of Observational HIV Epidemiological Research Europe.
HIV-1-related Hodgkin lymphoma in the era of combination antiretroviral therapy: incidence and evolution of CD4+ T-cell lymphocytes
.
Blood
.
2011
;
117
(
23
):
6100
-
6108
.
40.
Kowalkowski
MA
,
Mims
MP
,
Amiran
ES
,
Lulla
P
,
Chiao
EY
.
Effect of immune reconstitution on the incidence of HIV-related Hodgkin lymphoma [published correction appears in PLoS One. 2013;8(10): 10.1371/annotation/750f93e7-3fee-4ae0-81e4-058787a9e9a1]
.
PLoS One
.
2013
;
8
(
10
):
e77409
.
41.
Gotti
D
,
Danesi
M
,
Calabresi
A
, et al
; Brescia HIV Cancer Study Group.
Clinical characteristics, incidence, and risk factors of HIV-related Hodgkin lymphoma in the era of combination antiretroviral therapy
.
AIDS Patient Care STDS
.
2013
;
27
(
5
):
259
-
265
.
42.
Westhoff Smith
D
,
Sugden
B
.
Potential cellular functions of Epstein-Barr nuclear antigen 1 (EBNA1) of Epstein-Barr Virus
.
Viruses
.
2013
;
5
(
1
):
226
-
240
.
43.
Frappier
L
.
Contributions of Epstein-Barr nuclear antigen 1 (EBNA1) to cell immortalization and survival
.
Viruses
.
2012
;
4
(
9
):
1537
-
1547
.
44.
Frappier
L
.
EBNA1 and host factors in Epstein-Barr virus latent DNA replication
.
Curr Opin Virol
.
2012
;
2
(
6
):
733
-
739
.
45.
Frappier
L
.
The Epstein-Barr virus EBNA1 protein
.
Scientifica (Cairo)
.
2012
;
2012
:
438204
.
46.
Flavell
JR
,
Baumforth
KR
,
Wood
VH
, et al
.
Down-regulation of the TGF-beta target gene, PTPRK, by the Epstein-Barr virus encoded EBNA1 contributes to the growth and survival of Hodgkin lymphoma cells
.
Blood
.
2008
;
111
(
1
):
292
-
301
.
47.
Wood
VH
,
O’Neil
JD
,
Wei
W
,
Stewart
SE
,
Dawson
CW
,
Young
LS
.
Epstein-Barr virus-encoded EBNA1 regulates cellular gene transcription and modulates the STAT1 and TGFbeta signaling pathways
.
Oncogene
.
2007
;
26
(
28
):
4135
-
4147
.
48.
Lam
N
,
Sugden
B
.
CD40 and its viral mimic, LMP1: similar means to different ends
.
Cell Signal
.
2003
;
15
(
1
):
9
-
16
.
49.
Henderson
S
,
Rowe
M
,
Gregory
C
, et al
.
Induction of bcl-2 expression by Epstein-Barr virus latent membrane protein 1 protects infected B cells from programmed cell death
.
Cell
.
1991
;
65
(
7
):
1107
-
1115
.
50.
Vockerodt
M
,
Morgan
SL
,
Kuo
M
, et al
.
The Epstein-Barr virus oncoprotein, latent membrane protein-1, reprograms germinal centre B cells towards a Hodgkin’s Reed-Sternberg-like phenotype
.
J Pathol
.
2008
;
216
(
1
):
83
-
92
.
51.
Cahir-McFarland
ED
,
Carter
K
,
Rosenwald
A
, et al
.
Role of NF-kappa B in cell survival and transcription of latent membrane protein 1-expressing or Epstein-Barr virus latency III-infected cells
.
J Virol
.
2004
;
78
(
8
):
4108
-
4119
.
52.
Bargou
RC
,
Emmerich
F
,
Krappmann
D
, et al
.
Constitutive nuclear factor-kappaB-RelA activation is required for proliferation and survival of Hodgkin’s disease tumor cells
.
J Clin Invest
.
1997
;
100
(
12
):
2961
-
2969
.
53.
Dutton
A
,
Reynolds
GM
,
Dawson
CW
,
Young
LS
,
Murray
PG
.
Constitutive activation of phosphatidyl-inositide 3 kinase contributes to the survival of Hodgkin’s lymphoma cells through a mechanism involving Akt kinase and mTOR
.
J Pathol
.
2005
;
205
(
4
):
498
-
506
.
54.
Heath
E
,
Begue-Pastor
N
,
Chaganti
S
, et al
.
Epstein-Barr virus infection of naïve B cells in vitro frequently selects clones with mutated immunoglobulin genotypes: implications for virus biology
.
PLoS Pathog
.
2012
;
8
(
5
):
e1002697
.
55.
Holtick
U
,
Vockerodt
M
,
Pinkert
D
, et al
.
STAT3 is essential for Hodgkin lymphoma cell proliferation and is a target of tyrphostin AG17 which confers sensitization for apoptosis
.
Leukemia
.
2005
;
19
(
6
):
936
-
944
.
56.
Murray
P
,
Bell
A
.
Contribution of the Epstein-Barr virus to the pathogenesis of Hodgkin lymphoma
.
Curr Top Microbiol Immunol
.
2015
;
390
(
Pt 1
):
287
-
313
.
57.
Tiacci
E
,
Ladewig
E
,
Schiavoni
G
, et al
.
Pervasive mutations of JAK-STAT pathway genes in classical Hodgkin lymphoma
.
Blood
.
2018
;
131
(
22
):
2454
-
2465
.
58.
Jungnickel
B
,
Staratschek-Jox
A
,
Bräuninger
A
, et al
.
Clonal deleterious mutations in the IkappaBalpha gene in the malignant cells in Hodgkin’s lymphoma
.
J Exp Med
.
2000
;
191
(
2
):
395
-
402
.
59.
Schumacher
MA
,
Schmitz
R
,
Brune
V
, et al
.
Mutations in the genes coding for the NF-κB regulating factors IκBα and A20 are uncommon in nodular lymphocyte-predominant Hodgkin’s lymphoma
.
Haematologica
.
2010
;
95
(
1
):
153
-
157
.
60.
Schmitz
R
,
Hansmann
ML
,
Bohle
V
, et al
.
TNFAIP3 (A20) is a tumor suppressor gene in Hodgkin lymphoma and primary mediastinal B cell lymphoma
.
J Exp Med
.
2009
;
206
(
5
):
981
-
989
.
61.
Montgomery
ND
,
Coward
WB
IV
,
Johnson
S
, et al
.
Karyotypic abnormalities associated with Epstein-Barr virus status in classical Hodgkin lymphoma
.
Cancer Genet
.
2016
;
209
(
9
):
408
-
416
.
62.
Hu
LF
,
Chen
F
,
Zheng
X
, et al
.
Clonability and tumorigenicity of human epithelial cells expressing the EBV encoded membrane protein LMP1
.
Oncogene
.
1993
;
8
(
6
):
1575
-
1583
.
63.
Hu
LF
,
Zabarovsky
ER
,
Chen
F
, et al
.
Isolation and sequencing of the Epstein-Barr virus BNLF-1 gene (LMP1) from a Chinese nasopharyngeal carcinoma
.
J Gen Virol
.
1991
;
72
(
Pt 10
):
2399
-
2409
.
64.
Bellas
C
,
Santón
A
,
Manzanal
A
, et al
.
Pathological, immunological, and molecular features of Hodgkin’s disease associated with HIV infection. Comparison with ordinary Hodgkin’s disease
.
Am J Surg Pathol
.
1996
;
20
(
12
):
1520
-
1524
.
65.
Santón
A
,
Martín
C
,
Manzanal
AI
,
Preciado
MV
,
Bellas
C
.
Paediatric Hodgkin’s disease in Spain: association with Epstein-Barr virus strains carrying latent membrane protein-1 oncogene deletions and high frequency of dual infections
.
Br J Haematol
.
1998
;
103
(
1
):
129
-
136
.
66.
Zuercher
E
,
Butticaz
C
,
Wyniger
J
, et al
; Swiss HIV Cohort Study.
Genetic diversity of EBV-encoded LMP1 in the Swiss HIV Cohort Study and implication for NF-κΒ activation
.
PLoS One
.
2012
;
7
(
2
):
e32168
.
67.
Caldwell
RG
,
Wilson
JB
,
Anderson
SJ
,
Longnecker
R
.
Epstein-Barr virus LMP2A drives B cell development and survival in the absence of normal B cell receptor signals
.
Immunity
.
1998
;
9
(
3
):
405
-
411
.
68.
Merchant
M
,
Swart
R
,
Katzman
RB
, et al
.
The effects of the Epstein-Barr virus latent membrane protein 2A on B cell function
.
Int Rev Immunol
.
2001
;
20
(
6
):
805
-
835
.
69.
Chaganti
S
,
Bell
AI
,
Pastor
NB
, et al
.
Epstein-Barr virus infection in vitro can rescue germinal center B cells with inactivated immunoglobulin genes
.
Blood
.
2005
;
106
(
13
):
4249
-
4252
.
70.
Mancao
C
,
Altmann
M
,
Jungnickel
B
,
Hammerschmidt
W
.
Rescue of “crippled” germinal center B cells from apoptosis by Epstein-Barr virus
.
Blood
.
2005
;
106
(
13
):
4339
-
4344
.
71.
Bechtel
D
,
Kurth
J
,
Unkel
C
,
Küppers
R
.
Transformation of BCR-deficient germinal-center B cells by EBV supports a major role of the virus in the pathogenesis of Hodgkin and posttransplantation lymphomas
.
Blood
.
2005
;
106
(
13
):
4345
-
4350
.
72.
Mancao
C
,
Hammerschmidt
W
.
Epstein-Barr virus latent membrane protein 2A is a B-cell receptor mimic and essential for B-cell survival
.
Blood
.
2007
;
110
(
10
):
3715
-
3721
.
73.
Bräuninger
A
,
Schmitz
R
,
Bechtel
D
,
Renné
C
,
Hansmann
ML
,
Küppers
R
.
Molecular biology of Hodgkin’s and Reed/Sternberg cells in Hodgkin’s lymphoma
.
Int J Cancer
.
2006
;
118
(
8
):
1853
-
1861
.
74.
Portis
T
,
Dyck
P
,
Longnecker
R
.
Epstein-Barr Virus (EBV) LMP2A induces alterations in gene transcription similar to those observed in Reed-Sternberg cells of Hodgkin lymphoma
.
Blood
.
2003
;
102
(
12
):
4166
-
4178
.
75.
Portis
T
,
Longnecker
R
.
Epstein-Barr virus (EBV) LMP2A alters normal transcriptional regulation following B-cell receptor activation
.
Virology
.
2004
;
318
(
2
):
524
-
533
.
76.
Portis
T
,
Longnecker
R
.
Epstein-Barr virus LMP2A interferes with global transcription factor regulation when expressed during B-lymphocyte development
.
J Virol
.
2003
;
77
(
1
):
105
-
114
.
77.
Vockerodt
M
,
Wei
W
,
Nagy
E
, et al
.
Suppression of the LMP2A target gene, EGR-1, protects Hodgkin’s lymphoma cells from entry to the EBV lytic cycle
.
J Pathol
.
2013
;
230
(
4
):
399
-
409
.
78.
Sueur
C
,
Lupo
J
,
Germi
R
, et al
.
Characterization of Epstein-Barr Virus LMP1 deletion variants by next-generation sequencing in HIV-associated Hodgkin Lymphoma (French ANRS CO16 LYMPHOVIR cohort)
.
J Clin Virol
.
2016
;
82
:
S89
-
S90
.
79.
Kis
LL
,
Takahara
M
,
Nagy
N
,
Klein
G
,
Klein
E
.
Cytokine mediated induction of the major Epstein-Barr virus (EBV)-encoded transforming protein, LMP-1
.
Immunol Lett
.
2006
;
104
(
1-2
):
83
-
88
.
80.
Dukers
DF
,
Jaspars
LH
,
Vos
W
, et al
.
Quantitative immunohistochemical analysis of cytokine profiles in Epstein-Barr virus-positive and -negative cases of Hodgkin’s disease
.
J Pathol
.
2000
;
190
(
2
):
143
-
149
.
81.
Baumforth
KR
,
Birgersdotter
A
,
Reynolds
GM
, et al
.
Expression of the Epstein-Barr virus-encoded Epstein-Barr virus nuclear antigen 1 in Hodgkin’s lymphoma cells mediates up-regulation of CCL20 and the migration of regulatory T cells
.
Am J Pathol
.
2008
;
173
(
1
):
195
-
204
.
82.
Lee
SP
,
Constandinou
CM
,
Thomas
WA
, et al
.
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
.
1998
;
92
(
3
):
1020
-
1030
.
83.
Murray
PG
,
Constandinou
CM
,
Crocker
J
,
Young
LS
,
Ambinder
RF
.
Analysis of major histocompatibility complex class I, TAP expression, and LMP2 epitope sequence in Epstein-Barr virus-positive Hodgkin’s disease
.
Blood
.
1998
;
92
(
7
):
2477
-
2483
.
84.
Oudejans
JJ
,
Jiwa
NM
,
Kummer
JA
, et al
.
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
.
1996
;
87
(
9
):
3844
-
3851
.
85.
Reichel
J
,
Chadburn
A
,
Rubinstein
PG
, et al
.
Flow sorting and exome sequencing reveal the oncogenome of primary Hodgkin and Reed-Sternberg cells
.
Blood
.
2015
;
125
(
7
):
1061
-
1072
.
86.
Oudejans
JJ
,
Jiwa
NM
,
Kummer
JA
, et al
.
Activated cytotoxic T cells as prognostic marker in Hodgkin’s disease
.
Blood
.
1997
;
89
(
4
):
1376
-
1382
.
87.
Green
MR
,
Rodig
S
,
Juszczynski
P
, et al
.
Constitutive AP-1 activity and EBV infection induce PD-L1 in Hodgkin lymphomas and posttransplant lymphoproliferative disorders: implications for targeted therapy
.
Clin Cancer Res
.
2012
;
18
(
6
):
1611
-
1618
.
88.
Qiu
J
,
Cosmopoulos
K
,
Pegtel
M
, et al
.
A novel persistence associated EBV miRNA expression profile is disrupted in neoplasia
.
PLoS Pathog
.
2011
;
7
(
8
):
e1002193
.
89.
Lu
Y
,
Qin
Z
,
Wang
J
, et al
.
Epstein-Barr virus miR-BART6-3p inhibits the RIG-I Pathway
.
J Innate Immun
.
2017
;
9
(
6
):
574
-
586
.
90.
Tagawa
T
,
Albanese
M
,
Bouvet
M
, et al
.
Epstein-Barr viral miRNAs inhibit antiviral CD4+ T cell responses targeting IL-12 and peptide processing
.
J Exp Med
.
2016
;
213
(
10
):
2065
-
2080
.
91.
Albanese
M
,
Tagawa
T
,
Bouvet
M
, et al
.
Epstein-Barr virus microRNAs reduce immune surveillance by virus-specific CD8+ T cells
.
Proc Natl Acad Sci USA
.
2016
;
113
(
42
):
E6467
-
E6475
.
92.
Harold
C
,
Cox
D
,
Riley
KJ
.
Epstein-Barr viral microRNAs target caspase 3
.
Virol J
.
2016
;
13
(
1
):
145
.
93.
Vereide
DT
,
Seto
E
,
Chiu
YF
, et al
.
Epstein-Barr virus maintains lymphomas via its miRNAs
.
Oncogene
.
2014
;
33
(
10
):
1258
-
1264
.
94.
Ambrosio
MR
,
Navari
M
,
Di Lisio
L
, et al
.
The Epstein Barr-encoded BART-6-3p microRNA affects regulation of cell growth and immuno response in Burkitt lymphoma
.
Infect Agent Cancer
.
2014
;
9
(
1
):
12
.
95.
Office for National Statistics
.
Cancer survival in England–adults diagnosed
:
2009
to 2013, followed up to 2014. https://www.ons.gov.uk/peoplepopulationandcommunity/healthandsocialcare/conditionsanddiseases/bulletins/cancersurvivalinenglandadultsdiagnosed/2009to2013followedupto2014. Accessed 20 February 2019.
96.
Diepstra
A
,
van Imhoff
GW
,
Schaapveld
M
, et al
.
Latent Epstein-Barr virus infection of tumor cells in classical Hodgkin’s lymphoma predicts adverse outcome in older adult patients
.
J Clin Oncol
.
2009
;
27
(
23
):
3815
-
3821
.
97.
Jarrett
RF
,
Stark
GL
,
White
J
, et al
; Scotland and Newcastle Epidemiology of Hodgkin Disease Study Group.
Impact of tumor Epstein-Barr virus status on presenting features and outcome in age-defined subgroups of patients with classic Hodgkin lymphoma: a population-based study
.
Blood
.
2005
;
106
(
7
):
2444
-
2451
.
98.
Keegan
TH
,
Glaser
SL
,
Clarke
CA
, et al
.
Epstein-Barr virus as a marker of survival after Hodgkin’s lymphoma: a population-based study
.
J Clin Oncol
.
2005
;
23
(
30
):
7604
-
7613
.
99.
Clarke
CA
,
Glaser
SL
,
Dorfman
RF
, et al
.
Epstein-Barr virus and survival after Hodgkin disease in a population-based series of women
.
Cancer
.
2001
;
91
(
8
):
1579
-
1587
.
100.
Stark
GL
,
Wood
KM
,
Jack
F
,
Angus
B
,
Proctor
SJ
,
Taylor
PR
; Northern Region Lymphoma Group.
Hodgkin’s disease in the elderly: a population-based study
.
Br J Haematol
.
2002
;
119
(
2
):
432
-
440
.
101.
Meti
N
,
Esfahani
K
,
Johnson
NA
.
The role of immune checkpoint inhibitors in classical Hodgkin lymphoma
.
Cancers (Basel)
.
2018
;
10
(
6
):
E204
.
102.
Ansell
SM
,
Lesokhin
AM
,
Borrello
I
, et al
.
PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma
.
N Engl J Med
.
2015
;
372
(
4
):
311
-
319
.
103.
Younes
A
,
Santoro
A
,
Shipp
M
, et al
.
Nivolumab for classical Hodgkin’s lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial
.
Lancet Oncol
.
2016
;
17
(
9
):
1283
-
1294
.
104.
Green
MR
,
Monti
S
,
Rodig
SJ
, et al
.
Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma
.
Blood
.
2010
;
116
(
17
):
3268
-
3277
.
105.
Bollard
CM
,
Gottschalk
S
,
Torrano
V
, et al
.
Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins
.
J Clin Oncol
.
2014
;
32
(
8
):
798
-
808
.
106.
Rooney
CM
,
Smith
CA
,
Ng
CY
, et al
.
Infusion of cytotoxic T cells for the prevention and treatment of Epstein-Barr virus-induced lymphoma in allogeneic transplant recipients
.
Blood
.
1998
;
92
(
5
):
1549
-
1555
.
107.
Taylor
GS
,
Long
HM
,
Brooks
JM
,
Rickinson
AB
,
Hislop
AD
.
The immunology of Epstein-Barr virus-induced disease
.
Annu Rev Immunol
.
2015
;
33
(
1
):
787
-
821
.
108.
Ruhl
J
,
Citterio
C
,
Engelmann
C
, et al
.
Heterologous prime-boost vaccination protects from EBV antigen expressing lymphomas
.
J Clin Invest
.
2019
;
129
(
5
):
2071
-
2087
.
109.
Jiang
L
,
Xie
C
,
Lung
HL
, et al
.
EBNA1-targeted inhibitors: novel approaches for the treatment of Epstein-Barr virus-associated cancers
.
Theranostics
.
2018
;
8
(
19
):
5307
-
5319
.
110.
Thompson
S
,
Messick
T
,
Schultz
DC
,
Reichman
M
,
Lieberman
PM
.
Development of a high-throughput screen for inhibitors of Epstein-Barr virus EBNA1
.
J Biomol Screen
.
2010
;
15
(
9
):
1107
-
1115
.
111.
Lee
EK
,
Kim
SY
,
Noh
KW
, et al
.
Small molecule inhibition of Epstein-Barr virus nuclear antigen-1 DNA binding activity interferes with replication and persistence of the viral genome
.
Antiviral Res
.
2014
;
104
:
73
-
83
.
112.
Messick
TE
,
Smith
GR
,
Soldan
SS
, et al
.
Structure-based design of small-molecule inhibitors of EBNA1 DNA binding blocks Epstein-Barr virus latent infection and tumor growth
.
Sci Transl Med
.
2019
;
11
(
482
):
eaau5612
.
Sign in via your Institution