Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.

CCR-10-2578

Clinical
Cancer
Molecular Pathways Research

Epstein-Barr Virus–Associated B-cell Lymphomas: Pathogenesis


and Clinical Outcomes

Abhik Saha and Erle S. Robertson

Abstract
Epstein-Barr virus (EBV) is a ubiquitous human g-herpesvirus that establishes a life-long asymptomatic
infection in immunocompetent hosts. It is also found to be frequently associated with a broad spectrum of
B-cell lymphomas predominantly seen in immunodeficient patients. Despite many resemblances, these
EBV-linked lymphoproliferative disorders display heterogeneity at the clinical and the molecular level.
Moreover, EBV-associated lymphoproliferative diseases differ in their differential expression patterns of the
EBV-encoded latent antigens, which are directly related to their interactions with the host. EBV-driven
primary B-cell immortalization is linked to the cooperative functions of these latent proteins, which are
critical for perturbing many important cell-signaling pathways maintaining B-cell proliferation. Addition-
ally, it is used as a surrogate model to explore the underlying mechanisms involved in the development of
B-cell neoplasms. Recent discoveries have revealed that a number of sophisticated mechanisms are
exploited by EBV during cancer progression. This finding will be instrumental in the design of novel
approaches for therapeutic interventions against EBV-associated B-cell lymphomas. This review limits the
discussion to the biology and pathogenesis of EBV-associated B-cell lymphomas and the related clinical
implications. Clin Cancer Res; 17(10); 3056–63. 2011 AACR.

Background pattern of these latent genes defines the distinct latency


programs associated with the types of cancers (4). Type III
Epstein-Barr virus (EBV), a member of the g-herpesvirus latency, also referred to as the "growth program," expresses
family, is the first human tumor virus that was originally all the latent antigens and is typically found in EBV-asso-
identified in cultured lymphoblasts from Burkitt’s lym- ciated posttransplant lymphoproliferative diseases (PTLD),
phoma (BL) by Epstein and Barr in 1964 (1). Subsequent AIDS-associated lymphomas, and lymphoblastoid cell
studies showed that it is the causative agent of infectious lines (LCL; ref. 4). In vitro, EBV can infect primary B
mononucleosis, and >90% of the worldwide population is lymphocytes to generate continuously proliferating LCLs,
asymptomatically infected by EBV (2, 3). EBV infects both in which 4 of the viral latent antigens, EBNA-2, -3A, -3C,
B lymphocytes and epithelium cells, and intermittent reac- and LMP-1, are shown to be indispensable (4). Type II
tivation and virus replication in the oropharyngeal epithe- latency is characterized by a more restricted latent gene
lia allow the spreading of EBV infection and latent infection expression pattern (EBNA-1, LMP-1, -2), and is associated
in B lymphocytes (2, 3). with Hodgkin’s lymphoma (HL) and nasopharyngeal car-
During latent infection, to maintain the viral genome cinoma (NPC). Type I latency predominantly expresses
and to successfully evade the host cell immune surveil- EBNA1 and is associated with BL and gastric carcinoma
lance, EBV expresses a small subset of genes, including 6 (2, 5, 6).
nuclear antigens (EBNA-1, -2, -3A, -3B, -3C, and -LP), 3 One of the most significant features of EBV infection is
latent membrane proteins (LMP-1, -2A, and -2B), 2 small the engagement of different cell types and associated dis-
noncoding RNAs (EBER-1 and 2), and BamHI-A rightward eases (4). EBV is shown to be associated with a wide
transcripts (BART; refs. 2–4). The differential expression spectrum of human cancers, including both hematopoietic
and epithelial tumors, most preferentially in posttransplant
and AIDS patients (7). Despite its documented infection in
Authors' Affiliation: Department of Microbiology and Tumor Virology T lymphocytes and epithelial cells, EBV has a major pre-
Program, Abramson, Comprehensive Cancer Center, University of Penn- ference for B cells, and under certain circumstances the
sylvania Medical School, Philadelphia, Pennsylvania infected carrier B cells can transform into malignant B-cell
Corresponding Author: Erle S. Robertson, Department of Microbiology lymphomas (4). Because of the preferential infection in B
and Tumor Virology Program, Abramson, Comprehensive Cancer Center,
University of Pennsylvania Medical School, 201E Johnson Pavilion, 3610, cells, B-cell lymphomas are most prevalent among other
Hamilton Walk, Philadelphia, PA 19104. Phone: 215-746-0114; Fax: 215- EBV-associated lymphoproliferative disorders, which
746-0115; E-mail: erle@mail.med.upenn.edu include HL, BL, PTLDs, lymphomatoid granulomatosis,
doi: 10.1158/1078-0432.CCR-10-2578 senile EBV-associated B-cell lymphoproliferative disorders,
2011 American Association for Cancer Research. and many AIDS-associated B-cell lymphomas (4, 7). Given

3056 Clin Cancer Res; 17(10) May 15, 2011

Downloaded from clincancerres.aacrjournals.org on August 18, 2021. © 2011 American Association for Cancer
Research.
Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2578

EBV and B-Cell Lymphomas

the escalating list of EBV-associated human cancers, the heavy chain enhancer develop lymphomas (9). Further-
World Health Organization (WHO) classified EBV as a more, EBNA-1 may modulate its antiapoptotic function by
"carcinogenic agent" in 1997 (8). interacting with the host deubiquitinating enzyme HAUSP
Because of the critical association of EBV-associated (14). In response to DNA damage, HAUSP interacts with
lymphoproliferative diseases with viral latency, the func- and stabilizes p53. However, in EBV-infected cells, EBNA-1
tions of the latent antigens have been extensively studied. can efficiently block the interaction between HAUSP and
Nevertheless, further understanding the EBV life cycle, its p53, resulting in ubiquitin proteasome–mediated degrada-
genetic regulation, and the underlying molecular mechan- tion of p53 (5, 14). It should be noted that the effects of
isms are essential to fully comprehend the pathophysiology EBNA-1 on apoptosis are not only restricted to the regula-
of EBV infection in the spectrum of EBV-linked lympho- tion of p53 expression levels but also disrupt PML nuclear
proliferative syndromes and development of novel bodies in NPC cells (15) and lead to genomic instability by
therapies. Current knowledge has allowed us to infer that activating the recombinase-activating genes RAG-1 and
EBV-encoded latent antigens have evolved many sophisti- RAG-2 (9). Although it has long been thought that
cated strategies for manipulating important cellular path- EBNA-1–expressing cells escape from the cell-mediated
ways in the development of several human cancers (5). The immune system by blocking the MHC class I–restricted
important steps involved in EBV-mediated B-cell transfor- presentation, recent studies show that EBNA-1 can be
mation by the latent antigens are illustrated in Fig. 1, and presented to both CD4- and CD8-positive T cells; this
the biological functions of the latent proteins are discussed allows EBNA-1 to be used as a potential target for immu-
in detail below and summarized in Table 1. notherapy against EBV-associated tumors (2).
EBNA-2 plays a critical role during EBV-mediated host
Contribution of latent antigens to B-cell cell immortalization by coordinating the transcriptional
transformation level of viral and several cellular genes (3, 16). Together
EBNA-1 transcripts initiate from 4 different promoters: with EBNA-LP, EBNA-2 is the first EBV latent antigen
Wp, Cp, Qp, and Fp (2). EBNA-1 is transcribed from Wp detected after primary B-cell infection and suggested to
early during primary infection and switched over to Cp be involved in G0 to G1 phase transition (2). Because
after establishing the host cell transformation (2). During EBNA-2 does not bind DNA directly, the regulation of its
type I and II latency, Cp and Wp are epigenetically silenced, transcriptional activity largely depends on the interaction
and EBNA-1 transcription is initiated from Qp, which is with RBP-Jk (CBF1), a major Notch signaling mediator (2).
regulated in a cell cycle–dependent manner (2). Interest- EBNA-2 transactivates cellular genes, which include the B-
ingly, Fp is activated during the lytic cycle (2). EBNA-1 is cell activation markers CD21 and CD23, c-myc, hes-1, and
essential for replication and stable persistence of episomes runx3 (16). Viral genes that are activated by EBNA-2
in EBV-infected proliferating cells, and it is the only EBV- include LMP1, LMP2, and the Cp promoter, which regu-
encoded antigen that is consistently expressed in all EBV- lates transcription of EBNA genes (17). Two serologically
associated tumors (9). EBNA-1 contains 2 major functional distinct EBNA-2 proteins have been identified on the basis
domains: a C-terminal viral DNA-binding domain at OriP of significant sequence diversity in EBV types 1 and 2,
and an N-terminal host-chromosome–tethering domain. which also directly relates to functional consequences such
An X-ray crystallographic 3D structure of EBNA-1 reveals as B-cell transformation in vitro (3). Although EBNA-2 is
structural similarities with the functional homolog of essential for initial growth transformation of EBV-infected
human papillomavirus (HPV)–encoded E2 protein (10, B cells in vitro, its expression may be less critical during
11). Moreover, protein 3D structure prediction software cancer propagation as seen in most cases of EBV-linked
also indicated similar structural folds to another functional tumors, apart from those in immuno-suppressed patients
homolog, the Kaposi’s sarcoma associated Herpesvirus (2).
(KSHV)–encoded LANA protein (10, 11). This finding EBNA-LP (also known as EBNA-5) is concurrently
strongly suggests that EBNA-1 may serve as an attractive expressed with EBNA-2 in infected naive B cells (3).
therapeutic target for inhibition of viral latent replication Although a number of studies have suggested that
and persistence in EBV-associated tumors (11). In addition, EBNA-LP is important in B-cell growth transformation
increasing evidence suggests an oncogenic role for EBNA-1 through coactivation of EBNA-2–mediated transcriptional
in the progression of EBV-associated human cancers. For activity of the viral oncoprotein LMP1, the precise mechan-
example, downregulation of EBNA-1 expression using RNA istic details of its contribution remain elusive (2, 3). EBNA-
interference (RNAi) decreases cell proliferation and overall LP is an atypical viral protein comprised of a 22– and 44–
survival of many EBV-positive cancer cells (12). In agree- amino acid variable repeat region derived from 2 exons
ment with these observations, overexpression of a domi- (W1 and W2) found in the viral internal repeated region 1
nant-negative EBNA-1 mutant increases cell death in EBV- (IR1) and a unique 45-residue C terminus derived from the
positive BL cells, suggesting an antiapoptotic role for Y1 and Y2 exons, which gives rise to multiple protein
EBNA-1 (12). Furthermore, EBNA-1 expression increases species ranging from 20 to 130 kDa during initial infection
metastasis in nude mice through blocking the activity of because of the intrinsic number of repeats in the specific
metastasis suppressor protein Nm23–1H (13). In addition, viral genome in coordination with alternative splicing
mice carrying an EBNA-1 trans-gene under control of the Ig (2, 3). However, in vitro transformed LCLs express few

www.aacrjournals.org Clin Cancer Res; 17(10) May 15, 2011 3057

Downloaded from clincancerres.aacrjournals.org on August 18, 2021. © 2011 American Association for Cancer
Research.
Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2578

Saha and Robertson

D Host chromosome E
UbUb
HAUSP Ub
EBNA-1 EBNA1 p53 p53

Viral Degradation
episome
F EBNA-2
G EBNA-2 c-myc
EBNA-2 RBP-J␬ RBP-J␬ Cd21
LMP1 Cd23
Viral DNA Cellular DNA hes-1
runx3
A Viral entry
EBNA-LP
H EBNA-LP EBNA-2
RBP-J␬
LMP1
Viral DNA
I EBNA-2 J Mdm2
EBNA-3A E-3A E-3C p53
Oropharyngeal
epithelia EBNA-3B E-3B RBP-J␬ Ub Ub
Ub
EBNA-3C E-3C Blocks
B Modulates
p53
apoptosis
Notch signaling
L
Naive K CyclinD1
E-3C UbUb
B-cells E-3C CyclinA Ub
CDK6 Ub Ub P Facilitates CDK2 p27
C M Ub pRb G1-S transition SCFSkp2
SCFSkp2 E2F E2F
E-3C N E-3C
Latency III-Transformed B-cells
expressing all latent antigens
LMP-1
Q
Regulates JAK/STAT, ERK
LMP-1 MAPK, IRF, Wnt signaling

TRADD
O TRAF NF-␬B CD-23, -39, -40, -44,
bcl-2 HLAII, LFA-1, ICAM-1
Blocks P
apoptosis A20

LMP-2A LMP-2B
S
LMP-2A/2B

Lyn syk
Blocks BCR signaling
TBind to La, L22 R
EBERs Block apoptosis
Induce IFN, cytokines
U
BARTs ???

© 2011 American Association for Cancer Research

isoforms, and interestingly, its overall expression is quite dues, as a deletion of this particular domain resulted in
restricted in most of the EBV-associated tumors (2). Genetic viruses that immortalized B cells with less efficiency (2).
studies revealed the importance of the C-terminal 45 resi- Although EBNA-LP seems to be dispensable for growth

3058 Clin Cancer Res; 17(10) May 15, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on August 18, 2021. © 2011 American Association for Cancer
Research.
Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2578

EBV and B-Cell Lymphomas

transformation, recent biochemical studies have suggested contributes to the B-cell transformation (5, 6, 21, 22, 28,
that it may contribute to this process by critically regulating 29). Moreover, our group has shown that both EBNA-1 and
several important host components’ activity. EBNA-LP was -3C interact with the metastasis suppressor protein
shown to form complexes with major tumor suppressor Nm23–1H and positively modulate its NDP kinase and
proteins p53 and pRb, and cellular PKCs, such as HA95 and histidine kinase activities (5, 13). Furthermore, EBNA-3C,
HAX1, thereby facilitating the G0 to G1 phase transition of together with Nm23–1H, blocks Necdin-mediated growth
resting primary B cells after EBV infection (2, 18). suppression and antiangiogenic activities, suggesting their
The EBNA3 genes (-3A, -3B, and -3C) are in tandem critical role in cancer metastasis, and these antigens thus
sequence in the EBV genome and are transcribed as alter- could serve as potential candidates for prophylactic vacci-
nately spliced transcripts from the Cp promoter (19). These nation strategies (5).
nuclear proteins share approximately 30% sequence LMP1 is encoded by 3 exons and is an integral mem-
homology and are assumed to be derived from gene brane protein with 6 hydrophobic membrane-spanning
duplication events (2, 20). These proteins share more than segments and a C-terminal cytoplasmic section (2). Pos-
70% sequence homology between EBV type 1 and type 2 sibly through associating with the intermediate filament
strains, mainly because of variations in the C-terminal protein vimentin, LMP-1 forms patches in the plasma
repeat regions, and thus allowing strain typing (2). Reverse membrane (2). Subsequent mutational analyses showed
genetics have shown that EBNA-3B is dispensable, whereas that the conserved N-terminal and the first 2 trans-mem-
-3A and -3C are absolutely necessary for B-cell growth brane segments of LMP-1 are responsible for membrane
transformation along with EBNA-2 and LMP-1 (19). How- aggregation essential for B-cell immortalization (2). LMP-
ever, all 3 proteins affect global transcriptional activities 1 expression has been shown to have growth-transform-
including viral and several cellular gene expression patterns ing potential in rodent fibroblasts, as it promotes cellular
during host cell immortalization by modulating the alterations that are usually coupled with primary infec-
EBNA2/RBP-Jk–driven transactivation process (19, 20). tion and transformation of B cells. Moreover, in B cells,
EBNA-3A, together with EBNA-3C, facilitates LCL out- LMP-1 expression blocks p53-mediated apoptosis via
growth through epigenetic repression of the proapoptotic transactivation of the antiapoptotic factors bcl-2 and
protein Bim and the cell cycle inhibitor p16INK4A gene A20 (30). Interestingly, in agreement with this observa-
expression (19). EBNA-3C upregulates cellular CD21 tion, these genes are also shown to be upregulated in
and c-myc expression and augments EBNA2-driven upre- LMP-1 transgenic mice (31). In addition, LMP-1 upregu-
gulation of LMP1 expression and downregulates Cp-pro- lates interleukin (IL)–10 expression, which stimulates B-
moter activity (2, 21). In addition to their role in cell proliferation and reduces immune response (32).
transcriptional regulation, EBNA-3A and -3C also play Most importantly, LMP-1 augments host cell growth by
an important role in cell-cycle regulation. EBNA-3C resem- mimicking the CD40–mediated NF-kB signaling path-
bles adenovirus E1A and HPV E7 proteins, as its expression way, via interaction with TNF receptor (TNFR)–associated
efficiently overrides various cell-cycle checkpoints in factors (TRAF) and the TNFR-associated death domain
response to genotoxic stress through blocking p53-depen- protein (TRADD; ref. 33). LMP-1 interaction with TRAF-1
dent apoptotic activity (22, 23), as well as by escalating and -3 leads to elevated expression of many B-cell activa-
ubiquitin proteasome–mediated degradation of pRb and tion markers, including CD23, CD39, CD40, CD44,
p27KIP1 proteins (24–26). Besides, both EBNA-3A and -3C human leukocyte antigen (HLA) class II, and the cellular
cooperate with oncogenic H-ras in transforming primary adhesion components LFA-1 and ICAM-1 (33). More-
rat embryonic fibroblasts (5, 27). Studies from our labora- over, LMP-1 modulates Janus activated kinase (JAK)/
tory have shown that EBNA-3C interacts with a wide range STAT, extracellular signal regulated kinase (ERK) mitogen
of cellular proteins involved in cell cycle, apoptosis, and activated protein kinase (MAPK), IRF, and Wnt signaling
epigenetic transcriptional regulations, such as cyclin A, pathways (5, 33). These cell-signaling activities are
cyclin D1, SCFSkp2, pRb, c-Myc, p53, ING4, ING5, mediated by the 3 cytoplasmic C-terminal activating
Mdm2, p300, and histone deacetylase 1 (HDAC1), which regions (CTAR-1, -2, and -3) of LMP-1 (2, 33). LMP-1

Figure 1. The salient features of EBV-latent antigens in B-cell lymphomas. A, after initial infection of oropharyngeal epithelial cells, B, EBV primarily infects
naïve B cells; and, C, subsequently, the infected B cells are growth transformed (latency III), expressing the full repertoire of latent antigens. D, EBNA-1 tethers
viral episome to host chromosome and assists viral replication and segregation. E, EBNA-1 blocks the interaction between p53 and HAUSP and induces
its degradation. F, EBNA-2 regulates a wide range of both viral and, G, cellular gene transcription via RBP-Jk. H, EBNA-LP promotes EBNA-2–mediated gene
transcription. I, EBNA-3 proteins (-3A, -3B, and -3C), by blocking EBNA-2 association with RBP-Jk, modulate viral gene transcription as well as Notch
signaling pathway. J, EBNA-3C forms a ternary complex with p53 and Mdm2, recruits the Mdm2 E3 ligase activity to ease its degradation, and blocks
p53-dependent apoptosis. K, EBNA-3C binds to and enhances the kinase activity of both cyclin D1/CDK6 and, L, cyclin A/CDK2 complexes to phosphorylate
pRb. In addition, M, EBNA-3C recruits SCFSkp2 E3 ligase activity on hyperphosphorylated pRb and, N, p27KIP1 for degradation, thereby facilitating the
transition of G1 to S phase. O, LMP-1 inhibits p53-mediated apoptosis by transactivating bcl-2 and A20 genes. P, LMP-1 augments NF-kB signaling to
transactivate numerous cellular genes via interaction with TRAFs and TRADD. Q, LMP-1 also modulates JAK/STAT, ERK MAPK, IRF, and Wnt signaling
pathways. R, LMP-2A blocks BCR signaling through interaction with cellular tyrosine kinases Lyn and Syk. S, LMP-2B regulates LMP-2A function. T, EBERs
block apoptosis, bind to La and L22 cellular proteins, and induce IL-10 cytokine and type-I IFN. U, the biological function of BARTs and, specifically, their role
in development of B-cell lymphoma is under investigation.

www.aacrjournals.org Clin Cancer Res; 17(10) May 15, 2011 3059

Downloaded from clincancerres.aacrjournals.org on August 18, 2021. © 2011 American Association for Cancer
Research.
Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2578

Saha and Robertson

Table 1. Proposed Functions of EBV Latent Antigens and the Associated B-cell Lymphomas

Latent proteins Functions Associated lymphomas Latency type

EBNA-1 Essential for B-cell immortalization, BL I, II, III


replicates EBV genome, segregates Hodgkin's disease
viral episomes at mitosis, blocks AIDS-associated lymphomas
interaction between HAUSP Posttransplant
and p53 to facilitate p53's lymphoproliferative
degradation disorders
EBNA-2 Transcriptional coactivator that AIDS-associated lymphomas III
upregulates expression of viral Posttransplant
(LMP1) and cellular genes (c-myc), lymphoproliferative
essential for B-cell immortalization disorders
EBNA-3A Essential for B-cell immortalization AIDS-associated lymphomas III
of cell, interacts with RBP-Jk, regulates Posttransplant
Notch-signaling pathway lymphoproliferative
disorders
EBNA-3B Not essential for B-cell immortalization, AIDS-associated lymphomas III
interacts with RBP-Jk Posttransplant
lymphoproliferative
disorders
EBNA-3C Essential for B-cell immortalization, AIDS-associated lymphomas III
overcomes various checkpoints Posttransplant
in cell cycle, interacts with RBP-Jk, lymphoproliferative
activates LMP1, blocks p53-dependent disorders
apoptosis, enhances kinase activity
of both cyclin D1/CDK6 and cyclin
A/CDK2 complexes, induces degradation
p27KIP1 and pRb, stabilizes c-Myc,
Cyclin D1, and Mdm2, manipulates
host chromatin
remodeling machinery
EBNA-LP Interacts with EBNA2 to inactivate AIDS-associated lymphomas III
p53 and pRb, interacts with transcription Posttransplant
factors in notch signaling pathway, lymphoproliferative
contributes to B-cell immortalization disorders
LMP-1 Mimics CD40 ligand-binding signal, Hodgkin's disease II, III
stimulates bcl-2 and a20 expression AIDS-associated lymphomas
to block apoptosis, acts as a constitutively Posttransplant
active receptor for stimulating many cellular lymphoproliferative
genes, regulates NF-kB, JAK/STAT, ERK disorders
MAPK, IRF, and Wnt signaling pathways,
essential for B-cell immortalization
LMP-2A and -2B Drives EBV into latency, LMP-2A Hodgkin's disease II, III
blocks BCR signaling and -2B AIDS-associated lymphomas
assists its function, not essential Posttransplant
for B-cell immortalization lymphoproliferative
disorders
EBERs Form complexes with La and L22, BL I, II, III
associate with PKR, induce IFN and Hodgkin's disease
IL-10, bind to RIG-1 to activate AIDS-associated lymphomas
type I IFNs, not essential for B-cell Posttransplant lymphoproliferative
immortalization disorders
BARFs Protein products may modify Notch BL I, II, III
signaling, not essential for Hodgkin's disease
B-cell immortalization AIDS-associated lymphomas
Posttransplant lymphoproliferative
disorders

3060 Clin Cancer Res; 17(10) May 15, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on August 18, 2021. © 2011 American Association for Cancer
Research.
Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2578

EBV and B-Cell Lymphomas

can also affect the biogenesis of both cytokine and cyto- B-cell lymphomas remains largely inadequate. However,
kine receptor, which further alters overall angiogenesis recent studies have suggested that combined therapeutic
and inflammatory responses, thereby contributing to approaches of specific antiviral compounds targeting spe-
immune escape and cancer propagation (2, 3). cific viral antigens, with either T-cell–based immunother-
LMP-2 gene encodes 2 isoforms of the hydrophobic apy or targeted monoclonal antibodies, show promising
integral membrane protein, LMP-2A and -2B, which are outcomes against EBV-associated tumors.
transcribed from 2 distinct promoters after circularization In order to stimulate the host immune responses as a
of the EBV genome (2, 3). Similar to LMP-1, they are therapeutic strategy against EBV-associated B-cell lympho-
comprised of trans-membrane domains and a hydrophilic mas, efforts have been made in patients who have devel-
C-terminal domain (2, 3). In comparison to LMP-2B, oped PTLD after allogeneic hematopoietic stem cell
LMP-2A contains an extra 118-residue cytoplasmic N- transplantation (HSCT; refs. 3, 38). Adoptive immunother-
terminal domain encoded in exon 1, which interacts with apy using EBV-specific cytotoxic T lymphocytes (CTL) has
the cellular tyrosine kinases Lyn and Syk to block B-cell also been shown to be particularly useful in this context.
receptor (BCR) signaling, and also prevents lytic-cycle The CTLs can be generated from a healthy donor and
induction (34). Although LMP-2 is not required for B- infused directly into the recipient patient to reinstate
cell transformation, studies have clearly suggested that it immuno-competence (3). The infusion of unmanipulated
is essential for long-term persistence for the viral episome donor cells resulted in high response rates, although there
by providing temporal growth and B-cell survival signals is a considerable risk of graft-versus-host disease (GVHD)
in the lymphoid organs (3). LMP-2A seems to play a due to alloreactive CTLs (3). These aforementioned B-cell–
central role in the persistent infection of B cells. Recent related lymphomas represent an attractive target for CTL-
findings suggest that LMP-2B modulates LMP-2A function based immunotherapy, as the transformed B cells express
on regulating BCR signaling and thus driving the latently the full array of latent antigens. Moreover, the potential for
infected B cells to lytic reactivation (2, 3). success using this approach in clinical trial is facilitated by
Among 2 EBV nonpolyadenylated RNAs (EBER), EBER-1 the relative ease to obtain EBV-specific CTLs in large num-
is abundantly expressed, although in some instances, bers using in vitro transformed EBV-positive LCLs (3). In
EBER-2 is expressed at an elevated level (2). They interact patients with solid organ transplants (SOT) who develop
with cellular autoantigen La and the ribosomal protein L22 EBV-associated lymphoproliferative syndromes, EBV-spe-
(35). In addition, they block apoptosis, suppress antiviral cific CTLs have shown restricted response as the tumor
effects of IFN-a and -g, and induce IL-10 production, develops in recipient B cells (39). SOT patients require
perhaps mediated by binding to and blocking the function continuous immuno-suppression to prevent graft rejec-
of the IFN-induced double-stranded RNA-activated protein tion. Although adoptive transfer of EBV-specific CTLs
kinase PKR (2). Recently, EBERs have been shown to bind may help in restoring immuno-competence, CTL function
RIG-I and activate its downstream signaling, which further is still partially impaired by the immuno-suppressive drugs.
activates type-I IFNs (36). Moreover, EBERs can induce IL- Currently, strategies to genetically modify EBV-specific
10 via NF-kB–independent IRF3 activation in BL cells (37). CTLs for resistance against immuno-suppressive drugs
Although it was initially suggested that EBERs are not are been developed (40). Generation of EBV-specific CTLs
essential for EBV-mediated cell growth transformation, in this patient population offers various challenges com-
they may actively participate in cancer progression via pared with HSCT recipients, as the SOT donor is not HLA
modulation of the host-immune signaling activities (37). matched and usually deceased (40). Several groups have
EBV was the first human virus shown to encode micro- successfully administered autologous EBV-specific CTLs as
RNAs that mapped to the lytic BHRF1 and latent BART prophylaxis in SOT recipients (40, 41). However, making
regions of the genome (3). BARTs are transcribed from the autologous CTLs is sometimes not feasible. Consequently,
Bam H1A region of the viral episome (3). Although these a panel of CTLs is typically generated from healthy donors,
transcripts can be detected in various EBV-associated and only the HLA-matched CTLs are used for the treatment
tumors, they are expressed at elevated levels in NPCs (42). In contrast, limited clinical studies document EBV-
(3). Several open reading frames have been identified CTLs for other EBV-linked lymphomas and are mostly
within the differently spliced BARTs (3). The function of related to HL with a partial response because of alloreactive
most of the BARTs is still under investigation, but their CTLs (43). In addition, to protect against initial infection or
detection in infected B cells and in many established EBV- to enhance adoptive immunity in patients with EBV-asso-
associated lymphomas suggests that they might have an ciated neoplasm, the strategies would be to develop vac-
important role in viral persistence and pathogenesis. cines using combinations of several defined EBV epitopes
or linearly joined epitopes, expressed as chimeric polypep-
Clinical-Translational Advances tides to stimulate EBV-specific CTL immunity (44).
To date, several antiviral agents have shown encourag-
Despite current molecular advancements in exploring ing anti-EBV activity. However, most of these drugs dis-
the underlying mechanisms in EBV latent infection asso- play broad-spectrum activity against other viruses and
ciated with the development of its related diseases, the show limited efficacy against EBV-latent infection (2).
optimal management in controlling many EBV-associated For instance, drugs such as acyclovir and ganciclovir are

www.aacrjournals.org Clin Cancer Res; 17(10) May 15, 2011 3061

Downloaded from clincancerres.aacrjournals.org on August 18, 2021. © 2011 American Association for Cancer
Research.
Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2578

Saha and Robertson

nucleoside analogs that efficiently target the viral thymi- factors that critically modulate lytic-cycle mode will allow
dine kinase (TK), important for EBV lytic replication (45). us to evaluate the clinical potential for such a strategy.
In contrast, in latent EBV-associated B-cell lymphomas in Moreover, therapeutic advancements based on drugs, such
which TK is not expressed, antiviral therapy would be as hydroxyurea, capable of eliminating the viral episome
ineffective (45). To overcome this obstacle, ganciclovir is from tumor cells, may have clinical significance (38).
administered together with arginine butyrate, which Strategies undertaken with RNAi have been largely accepted
induces lytic cycle and TK production, and this has shown for the treatment of many virus-mediated cancers, in which
positive results in patients with PTLD (38). Similarly, other RNAi designed against viral latent antigens crucial for
chemotherapy agents may also induce lytic infection, and transformation, such as LMP1, have antitumor effects with-
1 study has shown that the combination of gemcitabine out altering the physiology of the cellular networks (38, 52,
and/or doxorubicin with ganciclovir is highly effective for 53). Recently, we also showed that a knockdown strategy
the inhibition of EBV-driven lymphoproliferation in severe for the EBNA3C gene efficiently blocks LCL and induces
combined immunodeficient mice (46). The viral DNA apoptosis, leading to the enhancement of the therapeutic
polymerase is another target for other antiviral compounds window against B-cell lymphoma (29). In addition, a
such as foscarnet and cidofovir, which show remarkable number of eccentric antiviral agents, including plant fla-
antitumor effects in patients (47). Interestingly, rituximab, vanoids, have also shown a variable degree of anti-EBV
a monoclonal antibody against CD20, either alone or activity (54). Thus, the future of therapeutic developments
coupled with cidofovir, eradicates PTLD symptoms in against EBV-associated B-cell lymphomas shows a great
organ-transplant patients with a success rate of more than deal of promise, which should translate into enhancing
60% (48, 49). Moreover, an HDAC inhibitor azelaic bishy- patient care.
droxamic acid and a retroviral agent zidovudine have been
used effectively against EBV-transformed B lymphocytes Disclosure of Potential Conflicts of Interest
from AIDS patients (50, 51). Several other virus-targeted
No potential conflicts of interest were disclosed.
therapeutic approaches are currently under consideration.
One such strategy is based on the induction of the EBV lytic Received January 19, 2011; accepted February 14, 2011; published
cycle (45). A better understanding of the viral and cellular OnlineFirst March 3, 2011.

References
1. Epstein MA, Achong BG, Barr YM. Virus particles in cultured lympho- 13. Murakami M, Kaul R, Kumar P, Robertson ES. Nucleoside dipho-
blasts from Burkitt's lymphoma. Lancet 1964;1:702–3. sphate kinase/Nm23 and Epstein-Barr virus. Mol Cell Biochem 2009;
2. Middeldorp JM, Brink AA, Van Den Brule AJ, Meijer CJ. Pathogenic 329:131–9.
roles for Epstein-Barr virus (EBV) gene products in EBV-associated 14. Saridakis V, Sheng Y, Sarkari F, Holowaty MN, Shire K, Nguyen T,
proliferative disorders. Crit Rev Oncol Hematol 2003;45:1–36. et al. Structure of the p53 binding domain of HAUSP/USP7 bound to
3. Thompson MP, Kurzrock R. Epstein-Barr virus and cancer. Clin Epstein-Barr nuclear antigen 1 implications for EBV-mediated immor-
Cancer Res 2004;10:803–21. talization. Mol Cell 2005;18:25–36.
4. Bajaj BG, Murakami M, Robertson ES. Molecular biology of EBV in 15. Sivachandran N, Sarkari F, Frappier L. Epstein-Barr nuclear antigen 1
relationship to AIDS-associated oncogenesis. Cancer Treat Res contributes to nasopharyngeal carcinoma through disruption of PML
2007;133:141–62. nuclear bodies. PLoS Pathog 2008;4:e1000170.
5. Saha A, Kaul R, Murakami M, Robertson ES. Tumor viruses and 16. Zimber-Strobl U, Strobl LJ. EBNA2 and Notch signalling in Epstein-
cancer biology: Modulating signaling pathways for therapeutic inter- Barr virus mediated immortalization of B lymphocytes. Semin Cancer
vention. Cancer Biol Ther 2010;10:961–78. Biol 2001;11:423–34.
6. Saha A, Murakami M, Kumar P, Bajaj B, Sims K, Robertson ES. 17. Schlee M, Schuhmacher M, Ho € lzel M, Laux G, Bornkamm GW. c-MYC
Epstein-Barr virus nuclear antigen 3C augments Mdm2-mediated impairs immunogenicity of human B cells. Adv Cancer Res 2007;97:
p53 ubiquitination and degradation by deubiquitinating Mdm2. J Virol 167–88.
2009;83:4652–69. 18. Klein G, Klein E, Kashuba E. Interaction of Epstein-Barr virus (EBV)
7. Ku €ppers R. Mechanisms of B-cell lymphoma pathogenesis. Nat Rev with human B-lymphocytes. Biochem Biophys Res Commun 2010;
Cancer 2005;5:251–62. 396:67–73.
8. Delecluse HJ, Feederle R, O’Sullivan B, Taniere P. Epstein Barr virus- 19. White RE, Groves IJ, Turro E, Yee J, Kremmer E, Allday MJ. Extensive
associated tumours: an update for the attention of the working co-operation between the Epstein-Barr virus EBNA3 proteins in the
pathologist. J Clin Pathol 2007;60:1358–64. manipulation of host gene expression and epigenetic chromatin
9. Schulz TF, Cordes S. Is the Epstein-Barr virus EBNA-1 protein an modification. PLoS ONE 2010;5:e13979.
oncogen? Proc Natl Acad Sci U S A 2009;106:2091–2. 20. Anderton E, Yee J, Smith P, Crook T, White RE, Allday MJ. Two
10. Lu F, Wikramasinghe P, Norseen J, Tsai K, Wang P, Showe L, et al. Epstein-Barr virus (EBV) oncoproteins cooperate to repress expres-
Genome-wide analysis of host-chromosome binding sites for sion of the proapoptotic tumour-suppressor Bim: clues to the patho-
Epstein-Barr Virus Nuclear Antigen 1 (EBNA1). Virol J 2010;7: genesis of Burkitt's lymphoma. Oncogene 2008;27:421–33.
262. 21. Bajaj BG, Murakami M, Cai Q, Verma SC, Lan K, Robertson ES.
11. Li N, Thompson S, Schultz DC, Zhu W, Jiang H, Luo C, et al. Discovery Epstein-Barr virus nuclear antigen 3C interacts with and enhances
of selective inhibitors against EBNA1 via high throughput in silico the stability of the c-Myc oncoprotein. J Virol 2008;82:4082–
virtual screening. PLoS ONE 2010;5:e10126. 90.
12. Sivachandran N, Cao JY, Frappier L. Epstein-Barr virus nuclear 22. Yi F, Saha A, Murakami M, Kumar P, Knight JS, Cai Q, et al. Epstein-
antigen 1 Hijacks the host kinase CK2 to disrupt PML nuclear bodies. Barr virus nuclear antigen 3C targets p53 and modulates its transcrip-
J Virol 2010;84:11113–23. tional and apoptotic activities. Virology 2009;388:236–47.

3062 Clin Cancer Res; 17(10) May 15, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on August 18, 2021. © 2011 American Association for Cancer
Research.
Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2578

EBV and B-Cell Lymphomas

23. Saha A, Bamidele A, Murakami M, Robertson ES. EBNA3C attenuates 39. Milpied N, Vasseur B, Parquet N, Garnier JL, Antoine C, Quartier P,
the function of p53 through interaction with the inhibitor of growth et al. Humanized anti-CD20 monoclonal antibody (Rituximab) in post
family proteins, 4 and 5. J Virol 2010;85:2079–88. transplant B-lymphoproliferative disorder: a retrospective analysis on
24. Knight JS, Sharma N, Robertson ES. Epstein-Barr virus latent antigen 32 patients. Ann Oncol 2000;11[Suppl 1]:113–6.
3C can mediate the degradation of the retinoblastoma protein through 40. Vera JF, Brenner MK, Dotti G. Immunotherapy of human cancers
an SCF cellular ubiquitin ligase. Proc Natl Acad Sci U S A 2005;102: using gene modified T lymphocytes. Curr Gene Ther 2009;9:396–408.
18562–6. 41. Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adop-
25. Knight JS, Sharma N, Robertson ES. SCFSkp2 complex targeted by tive cell transfer: a clinical path to effective cancer immunotherapy.
Epstein-Barr virus essential nuclear antigen. Mol Cell Biol 2005;25: Nat Rev Cancer 2008;8:299–308.
1749–63. 42. Murray PG, Young LS. The role of the Epstein-Barr virus in human
26. Maruo S, Wu Y, Ito T, Kanda T, Kieff ED, Takada K. Epstein-Barr virus disease. Front Biosci 2002;7:d519–40.
nuclear protein EBNA3C residues critical for maintaining lymphoblas- 43. Gottschalk S, Heslop HE, Roon CM. Treatment of Epstein-Barr virus-
toid cell growth. Proc Natl Acad Sci U S A 2009;106:4419–24. associated malignancies with specific T cells. Adv Cancer Res 2002;
27. Hickabottom M, Parker GA, Freemont P, Crook T, Allday MJ. Two 84:175–201.
nonconsensus sites in the Epstein-Barr virus oncoprotein EBNA3A 44. Moss DJ, Schmidt C, Elliott S, Suhrbier A, Burrows S, Khanna R.
cooperate to bind the co-repressor carboxyl-terminal-binding protein Strategies involved in developing an effective vaccine for EBV-asso-
(CtBP). J Biol Chem 2002;277:47197–204. ciated diseases. Adv Cancer Res 1996;69:213–45.
28. West MJ. Structure and function of the Epstein-Barr virus transcription 45. Gershburg E, Pagano JS. Epstein-Barr virus infections: prospects for
factor, EBNA 3C. Curr Protein Pept Sci 2006;7:123–36. treatment. J Antimicrob Chemother 2005;56:277–81.
29. Saha A, Halder S, Upadhayay SK, Lu J, Kumar P, Murakami M, et al. 46. Heslop HE. Biology and treatment of Epstein-Barr virus-associated
EBNA3C facilitates G1-S transition by stabilizing and enhancing the non-Hodgkin lymphomas. Hematology (Am Soc Hematol Educ Pro-
function of Cyclin D1. PLoS Pathog 2010;7:e1001275. gram) 2005:260–6.
30. D’Souza BN, Edelstein LC, Pegman PM, Smith SM, Loughran ST, 47. De Clercq E. Antivirals and antiviral strategies. Nat Rev Microbiol
Clarke A, et al. Nuclear factor kappa B-dependent activation of the 2004;2:704–20.
antiapoptotic bfl-1 gene by the Epstein-Barr virus latent membrane 48. Kuehnle I, Huls MH, Liu Z, Semmelmann M, Krance RA, Brenner MK,
protein 1 and activated CD40 receptor. J Virol 2004;78:1800–16. et al. CD20 monoclonal antibody (rituximab) for therapy of Epstein-
31. Kulwichit W, Edwards RH, Davenport EM, Baskar JF, Godfrey V, Barr virus lymphoma after hemopoietic stem-cell transplantation.
Raab-Traub N. Expression of the Epstein-Barr virus latent membrane Blood 2000;95:1502–5.
protein 1 induces B cell lymphoma in transgenic mice. Proc Natl Acad 49. Ha€nel M, Fiedler F, Thorns C. Anti-CD20 monoclonal antibody (Ritux-
Sci U S A 1998;95:11963–8. imab) and Cidofovir as successful treatment of an EBV-associated
32. Lambert SL, Martinez OM. Latent membrane protein 1 of EBV acti- lymphoma with CNS involvement. Onkologie 2001;24:491–4.
vates phosphatidylinositol 3-kinase to induce production of IL-10. J 50. Sculley TB, Buck M, Gabrielli B, Parsons PG, Krauer KG. A histone
Immunol 2007;179:8225–34. deacetylase inhibitor, azelaic bishydroxamic acid, shows cytotoxicity
33. Graham JP, Arcipowski KM, Bishop GA. Differential B-lymphocyte on Epstein-Barr virus transformed B-cell lines: a potential therapy for
regulation by CD40 and its viral mimic, latent membrane protein 1. posttransplant lymphoproliferative disease. Transplantation 2002;73:
Immunol Rev 2010;237:226–48. 271–9.
34. Merchant M, Swart R, Katzman RB, Ikeda M, Ikeda A, Longnecker R, 51. Abdulkarim B, Bourhis J. Antiviral approaches for cancers related to
et al. The effects of the Epstein-Barr virus latent membrane protein 2A Epstein-Barr virus and human papillomavirus. Lancet Oncol 2001;
on B cell function. Int Rev Immunol 2001;20:805–35. 2:622–30.
35. Samanta M, Takada K. Modulation of innate immunity system by 52. Takeshita F, Ochiya T. Therapeutic potential of RNA interference
Epstein-Barr virus-encoded non-coding RNA and oncogenesis. Can- against cancer. Cancer Sci 2006;97:689–96.
cer Sci 2010;101:29–35. 53. Li XP, Li G, Peng Y, Kung HF, Lin MC. Suppression of Epstein-Barr
36. Samanta M, Iwakiri D, Kanda T, Imaizumi T, Takada K. EB virus- virus-encoded latent membrane protein-1 by RNA interference inhi-
encoded RNAs are recognized by RIG-I and activate signaling to bits the metastatic potential of nasopharyngeal carcinoma cells.
induce type I IFN. EMBO J 2006;25:4207–14. Biochem Biophys Res Commun 2004;315:212–8.
37. Iwakiri D, Takada K. Role of EBERs in the pathogenesis of EBV 54. Itoigawa M, Ito C, Ju-ichi M, Nobukuni T, Ichiishi E, Tokuda H, et al.
infection. Adv Cancer Res 2010;107:119–36. Cancer chemopreventive activity of flavanones on Epstein-Barr virus
38. Israel BF, Kenney SC. Virally targeted therapies for EBV-associated activation and two-stage mouse skin carcinogenesis. Cancer Lett
malignancies. Oncogene 2003;22:5122–30. 2002;176:25–9.

www.aacrjournals.org Clin Cancer Res; 17(10) May 15, 2011 3063

Downloaded from clincancerres.aacrjournals.org on August 18, 2021. © 2011 American Association for Cancer
Research.
Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2578

Epstein-Barr Virus−Associated B-cell Lymphomas:


Pathogenesis and Clinical Outcomes
Abhik Saha and Erle S. Robertson

Clin Cancer Res 2011;17:3056-3063. Published OnlineFirst March 3, 2011.

Updated version Access the most recent version of this article at:
doi:10.1158/1078-0432.CCR-10-2578

Cited articles This article cites 53 articles, 14 of which you can access for free at:
http://clincancerres.aacrjournals.org/content/17/10/3056.full#ref-list-1

Citing articles This article has been cited by 12 HighWire-hosted articles. Access the articles at:
http://clincancerres.aacrjournals.org/content/17/10/3056.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, use this link
http://clincancerres.aacrjournals.org/content/17/10/3056.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's
(CCC)
Rightslink site.

Downloaded from clincancerres.aacrjournals.org on August 18, 2021. © 2011 American Association for Cancer
Research.

You might also like