You are on page 1of 14

PERSPECTIVES

agent responsible for causing tumours


TIMELINE
in chickens, so was extremely receptive
to the somewhat unfashionable notion
Epstein–Barr virus: more than that viruses might play a role in the
development of human cancer. He had
50 years old and still providing the further advantage of having worked
on the application of electron microscopy
surprises to the study of RSV, having learned the
technology in the Rockefeller Institute
laboratory of George Pallade, who
Lawrence S. Young, Lee Fah Yap and Paul G. Murray subsequently won the Nobel Prize for his
development of electron microscopy to
Abstract | It is more than 50 years since the Epstein–Barr virus (EBV), the first human probe cellular ultrastructure. So it was that
tumour virus, was discovered. EBV has subsequently been found to be associated Epstein, by then working at the Middlesex
with a diverse range of tumours of both lymphoid and epithelial origin. Progress in Hospital, attended a lecture entitled ‘The
the molecular analysis of EBV has revealed fundamental mechanisms of more commonest children’s cancer in tropical
Africa: a hitherto unrecognized syndrome’,
general relevance to the oncogenic process. This Timeline article highlights key given by Burkitt on 22 March 1961. In
milestones in the 50‑year history of EBV and discusses how this virus provides a Epstein’s own words — “I was struck from
paradigm for exploiting insights at the molecular level in the diagnosis, treatment the outset by the highly unusual nature
and prevention of cancer. of the new tumour and by the exciting and
unprecedented findings on epidemiology
Approximately 2 million cases of cancer infection can result in EBV-associated B cell which seemed to show that its distribution
each year are caused by infectious agents1. tumours. The role of EBV in the pathogenesis depended on climatic factors indicating that
Studying the role of infectious agents in of epithelial tumours — namely, undifferen- some biological agent might be involved
cancer pathogenesis, in both humans and tiated nasopharyngeal carcinoma (NPC) and in the aetiology” (REF. 6). As Burkitt was
animals, has provided important insights into EBV-associated gastric carcinoma (EBV‑GC) talking, Epstein formulated the notion that a
the mechanisms that drive the oncogenic — remains unclear but is thought to result climate-dependent arthropod vector might
process. This understanding has also from the aberrant establishment of latent be involved in the spread of an oncogenic
identified opportunities for therapeutic viral infection in epithelial cells with existing virus. Although it later became clear that this
and prophylactic intervention. premalignant genetic changes4. Although the was not precisely the case, this idea marked
Epstein–Barr virus (EBV) was the first precise contribution of EBV to the oncogenic a crossroads in Epstein’s career and heralded
human tumour virus to be discovered. It is process is uncertain, the presence of the virus the advent of human tumour virology.
estimated that EBV accounts for more than in all tumour cells provides opportunities Epstein met Burkitt immediately after
200,000 cases of cancer each year and that for the development of novel therapeutic the lecture, and in discussions a few days
1.8% of all cancer deaths are due to EBV- interventions and diagnostic approaches. later Burkitt agreed to send fresh tumour
attributable malignancies1,2. The higher In this Timeline article, we provide an biopsies from Kampala for analysis
prevalence of EBV-associated tumours in the overview of the major milestones in EBV in Epstein’s laboratory using electron
developing world compared with developed research and highlight the insights that microscopy, along with more traditional
regions and the contribution of EBV to this virus continues to provide into cancer virus culture approaches, in an attempt
cancer development in immuno­suppressed pathogenesis and treatment (FIG. 1). to identify and rescue the putative virus
patients1 highlight important facets of the resident within Burkitt lymphoma (BL)
oncogenic process, including the role of The discovery of EBV cells3. Over almost 3 years none of these
multiple risk factors and the contribution In the context of the discovery of EBV it is attempts was successful and so Epstein, by
of loss of immune control. eminently appropriate that Louis Pasteur, then working with Yvonne Barr and Bert
EBV is the most common and father of germ theory, is also responsible for Achong, decided to attempt to culture the
persistent virus infection in humans, with a well-known aphorism — “chance favours BL cells. Epstein knew that certain chicken
approximately 95% of the world’s population the prepared mind” (REF. 5). For it was the tumour viruses only became evident when
sustaining an asymptomatic life-long ‘prepared mind’ of a young pathologist, they reproduced as a consequence of
infection3. This persistence is the result Anthony Epstein, and his ‘chance’ attendance reactivation induced by in vitro culture6.
of EBV’s unique interaction with B cells, at a talk by Denis Burkitt, a surgeon working However, in the early 1960s no one had
whereby the infection resides in the memory in Uganda, that led to the discovery of succeeded in growing any type of human
B cell pool in normal, healthy individuals. EBV6. From 1948 Epstein had worked on lymphocyte, despite innumerable attempts
Disruption of this finely regulated B cell Rous sarcoma virus (RSV), the transmissible using a variety of different approaches.

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

• VCA IgA levels detected in mass • EBV genome


serological screening of NPC patients detected in
ACIF developed to in China, confirming NPC risk166 ENK/T cell
detect EBNA1 protein • EBNA1 maintains the EBV genome212 lymphoma155
in BL and NPC cells56 • LMP1 is a classic oncogene221 • Latent gene
EBV particles EBV transformed EBV genome expression
discovered in lymphocytes in detected in EBV antibody and genome EBV genome in NPC
BL cells7 culture10,11 NPC cells169 detected in PTLD106 sequenced16 identified32

1964 1966 1967 1968 1970 1973 1974 1975 1978 1980 1982 1984 1985 1987 1988

• High EBV EBV EBV genome XLP1 was first described121 • HIV is a co-factor for • EBV DNA
antibody levels causes detected in EBV-positive BL149 detected in
in BL and NPC40 IM78 BL cells and Epidemiological study confirms • EBNA2 is essential for HL biopsies132
• EBV found in extracts link between EBV and BL44 B cell transformation215 • Latent gene
all human from NPC expression in
populations41 tumours55 • BZLF1 is the switch from latent to lytic cycle36 BL identified30
• EBV replication observed in oral ‘hairy’ leukoplakia80
• EBNA protein detected in HRS cells of a single HL patient131

Figure 1 | Timeline showing the major milestones in EBV research. ACIF, anti-complement immunofluorescence; BL, Burkitt lymphoma; EBER ISH,
Epstein–Barr virus-encoded RNA in situ hybridization; EBNA, Epstein–Barr nuclear antigen; EBV, Epstein–Barr virus; ENK, extranodal natural killer; HIV,
human immunodeficiency virus; HL, Hodgkin lymphoma; HRS, Hodgkin–Reed–Sternberg; IgA, immunoglobulin A; IM, infectious mononucleosis; LMP,
latent membrane protein; NPC, nasopharyngeal carcinoma; PTLD, post-transplant lympho­proliferative disease; Q‑PCR, quantitative PCR; TNFR, tumour
necrosis factor receptor; VCA, viral capsid antigen; XLP1, X‑linked lymphoproliferative disease type 1.

It is here that serendipity played a part: of EBV, Volker Diehl, working in the immortalization (FIG. 2). Subsequent genetic
a delayed and rerouted flight from Kampala laboratories of Werner and Gertrude studies revealed the existence of two EBV
in December 1963 resulted in a BL biopsy Henle, used an irradiated EBV producer types17,18 (BOX 1) as well as substantial genetic
reaching the London laboratory later than BL cell line (Jijoye) or a non-producer BL variation, affecting mainly the latent genes,
expected. The transit medium in which the cell line control (Raji) co‑cultivated with within each type19–29. The impact of genetic
BL biopsy was shipped was unusually cloudy sex-mismatched cord blood lymphocytes. variation on the biology of EBV infection
but, rather than the suspected contamination Within weeks, growth of the lymphocytes remains only poorly understood.
with bacteria, microscopic examination was observed only after co‑culture with Jijoye The EBV genome within LCLs usually
revealed viable tumour cells floating free of cells, confirming that this effect was most exists in multiple copies of extrachromo-
the main lymphoma mass. These cells grew likely due to EBV infection10. This study somal circular genetic material known as
when suspended in fresh culture medium, was published in 1967, the same year that episomes and expresses all latent genes
requiring the culture to be split into new John Pope in Brisbane had observed that cell (referred to as latency III or the ‘growth
culture bottles. And so the first cell line lines could readily be established from the programme’), including six Epstein–Barr
from a human lymphoma was established, peripheral blood lymphocytes of patients nuclear antigens (EBNAs 1, 2, 3A, 3B and
and early in 1964, Epstein prepared these with infectious mononucleosis (IM)11. The 3C and EBNA leader protein (EBNA‑LP)),
BL cells (now named the EB1 cell line after cell lines from patients with IM are the result latent membrane proteins LMP1 and LMP2
Epstein and Barr) for electron microscopy 3,6. of spontaneous transformation — the (which encodes two isoforms, LMP2A and
He was “exhilarated to observe unequivocal occasional growth of EBV-transformed LMP2B) and the non-coding EBV-encoded
virus particles in a cultured BL cell” (REF. 6) B cells from the blood of EBV-infected RNAs (EBER1 and EBER2) and viral
and it was clear from the morphology of the individuals. Pope then took filtrates from one microRNA (miRNA) (FIG. 2). During various
virus particles that this was a herpesvirus6. of these B cell lines and demonstrated that stages of B cell differentiation in vivo, EBV
Unlike the three other human herpesviruses they could efficiently transform fetal human expresses either the latency III programme,
known at that time, this virus was inert as lymphocytes12. These experiments confirmed or one of two alternative forms of virus
it did not induce the characteristic lytic that EBV was indeed a potent transforming latency (known as latency I and latency II).
infection usually associated with herpesvirus virus in cell culture. EBV remains the EBV-associated B cell lymphomas express
replication. These observations were most efficient transforming agent, rapidly either latency I, II or III depending, at
published in a Lancet paper 7 that became converting approximately 3–10% of all least partly, on the B cell stage from which
a citation classic in 1979 (REF. 8). target B cells into permanently growing each tumour is derived30,31. In contrast, all
lymphoblastoid cell lines (LCLs)13–15. EBV-associated epithelial cancers express
The first human tumour virus The publication in 1984 of the complete a latency II programme, or a variant
Discovering the potent transforming sequence of an EBV strain (the B95‑8 thereof 32,33. The contribution of these latent
ability of EBV. As more BL cell lines were strain)16 was the stimulus for a series of genes to the oncogenic process remains the
established9, the next key objective was to studies that identified individual viral subject of intense study (BOX 2).
determine whether EBV could transform genes, including the so‑called latent EBV is also able to induce its lytic cycle,
normal B lymphocytes. In the absence of genes, the coordinated expression of leading to virus replication. Although the
a BL cell line that produced large amounts which is responsible for in vitro B cell lytic cycle was known to be activated by

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

• NPC tumour cells carry


clonal EBV genomes170 • Complete
• LMP1 is essential • LMP1 is a constitutively sequences of 71
• EBER ISH for B cell activated member of Use of Q-PCR EBV strains
assays transformation222 the TNFR superfamily145 to detect Wild-type First controlled published29
developed134 • EBNA3A and 3C are • First successful use of cell-free EBV EBV trial of an EBV • Further
• LMP2 essential for B cell EBV-specific T cell DNA as NPC sequence vaccine (gp350 development of
identified224 transformation217 therapy in PTLD117 biomarker168 released19 vaccine)103 gp350 vaccine101

1989 1990 1992 1993 1994 1995 1998 1999 2002 2003 2004 2007 2012 2015

EBV genome • Identification of three HL is a B cell EBV Wp-restricted EBV EBNA3B is a


detected in forms of latency31 lymphoma135 persistence latency microRNAs tumour
HRS cells of • First demonstration of in memory identified in BL73 discovered232 suppressor219
HL biopsies133 EBV in typical gastric B cells in
adenocarcinomas180 vivo81

Nature Reviews | Cancer


diverse stimuli, including the phorbol ester loss of T cell control of EBV, both of which MYC oncogene on chromosome 8, placing
12‑O-tetra-decanoylphorbol‑13‑acetate could increase the pool of EBV-infected it under the transcriptional control of
(TPA) and sodium butyrate34,35, in 1985 BL progenitors45–48. an immunoglobulin locus62. The MYC
George Miller showed that the gene Although malaria impairs T cell translocations are also found in the
responsible for the latent to lytic cycle switch immunity to EBV, this impairment may be EBV-negative forms of BL that occur
was BZLF1 (REF. 36). Since then, the functions transient, whereas the increased EBV loads sporadically worldwide, and because they
of many of the virus genes expressed during in circulating memory B cells associated are required to sustain the high rate of
the lytic cycle have been characterized with malarial infection are long-lived45,49,50. proliferation characteristic of BL, MYC
(BOX 3). Although the precise in vivo Plasmodium falciparum, the parasite that abnormalities are now considered the
conditions responsible for activating the EBV causes malaria, may also act more directly hallmark of BL63,64.
lytic cycle remain unknown, this process is to induce BL. For example, it has been The demonstration that Akata BL cells
intimately related to the differentiation of shown that P. falciparum can increase that had lost EBV episomes in culture
both B cells and epithelial cells37–39. expression of the enzyme activation-­ were more sensitive to apoptosis than the
induced cytidine deaminase (AID), which parental virus-positive cells provided
Defining the aetiological role of EBV in induces BL‑causing mutations, including the first evidence that EBV might contribute
BL. Only 2 years after the discovery of the characteristic translocations51,52. Other an anti-apoptotic function that counteracts
EBV, Lloyd Old and colleagues40 described environmental factors may also be involved. MYC-driven apoptosis65–67. EBNA1 was
antibodies in the serum of African patients For example, in areas endemic for BL, initially shown to mediate some of the
with BL that recognized antigens from extracts of the plant Euphorbia tirucalli have anti-apoptotic effects of EBV in BL cells68.
BL cell lines. However, the first definitive many traditional uses; these extracts have However, EBNA1 alone is not sufficient
clues of a pathogenic role for EBV in been shown to induce the EBV lytic cycle to reconstitute complete protection from
BL came from sero-epidemiological and to induce chromosomal abnormalities cell death and other groups have shown an
studies that reported higher titres of EBV in B cell lines53,54. anti-apoptotic function for the EBERs in
antibodies in the serum of patients with More direct evidence of a causal BL69,70. The restricted pattern of virus latency
BL compared with age-matched controls relationship between EBV and BL was observed in BL is also apparently important
living in the same area41–43. The definitive provided in 1970 by Harald zur Hausen55, for the BL phenotype, as forced expression of
epidemiological study published in 1978 who used DNA hybridization to identify the full spectrum of latent genes in BL cells
was performed by Guy de Thé who the EBV genome in BL (and also in NPC; antagonizes MYC-driven oncogenesis71,72.
tested pre‑BL serum from around 42,000 see below). In 1974, Beverley Reedman56 Although most BLs express a so‑called
Ugandan children44. Of the 14 children who used anti-complement immunofluores- type I form of latency probably restricted
subsequently developed BL, most had raised cence (ACIF) to show, for the first time, to protein expression of EBNA1, Alan
EBV antibody titres long before diagnosis, expression of an EBV protein in BL cell Rickinson’s group73,74 showed that some BLs
leading to the conclusion that “BL develops lines; initially named ‘EBNA’, it is now express additional virus genes, including
in children who have had a long and heavy known as EBNA1 (REFS 57,58). EBNA3A, EBNA3B, EBNA3C and EBNA‑LP,
exposure to EBV” (REF. 44). Although this Work in the laboratories of George together with an EBNA2 deletion that
study failed to find marked differences in and Eva Klein showed that BL was suppresses the full latency programme.
the burden of malaria in BL cases compared characterized by abnormalities of These are often referred to as Wp‑restricted
with controls, the authors still regarded chromosome 14 (REF. 59), later shown to BL because virus gene expression is driven
malaria as an important cofactor, on the be the result of translocations involving from the Wp, rather than from the Qp,
basis of the tumour’s unique geographical chromosomes 14 and 8 (REF. 60). There are promoter (FIG. 2a). Wp‑restricted BL cell
distribution44. Malaria probably induces two less-common variant translocations61; lines are less sensitive to apoptosis than
an intense polyclonal B cell activation and all three deregulate expression of the conventional EBV-positive BL lines, an

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

a EBV genome: latent genes EBER1 effect attributed to downregulation of the


EBER2 pro-apoptotic molecule BIM (also known
LMP2A Wp
as BCL2L11) by EBNA3A and EBNA3C
LMP2B and to the overexpression of the viral BCL‑2
BARTs Cp
BARF1 homologue, BamHI fragment H rightward
open reading frame 1 (BHRF1), a gene
TR
LMP1
OriP originally thought to be expressed only in
the virus lytic cycle75,76. Recent global virus
gene expression studies indicate that BHRF1
is not the only lytic gene expressed in BL, at
Double-stranded
DNA episome least at the RNA level77.
EBNA-LP In this section we have described how the
EBNA2 discovery of EBV in BL eventually led to a
BHRF1 better understanding of the molecular basis
EBNA1
of this tumour. Below, we discuss how the
EBNA2 detection of EBV in other forms of cancer
deletion
led to the discovery of more unexpected
virus associations and of additional
Qp
EBNA3C mechanisms of virus-driven oncogenesis.
EBNA3B
EBNA3A Infections in asymptomatic hosts
Although the detection of antibodies to
EBV antigens in the sera of patients with
b Position of latent gene products on a linear genome BL provided the first evidence that EBV
Nhet OriP a S e1–e3 Z R c bT d might be causally linked to BL, antibodies
Nhet to EBV were also found to be present, albeit
CW W W W W W Y H F QU P O M L E K B G D XV I A
at lower levels, in normal healthy people,
TR W WWW W W EBNA1 TR including children41. A breakthrough in our
EBNA3A EBNA3C understanding of primary infection came
EBNA-LP EBNA3B LMP1 from another chance observation, this time
from the Henle laboratory, where a young
EBNA2 BHRF1
BARF1 technician, who was regularly bled as a
negative control in serological assays for EBV,
miR-BHRF1-1 miR-BHRF1-2 miR-BART-3 miR-BART-21 miR-BART-2
miR-BHRF1-3 miR-BART-4 miR-BART-18 became seropositive after developing IM,
miR-BART-1 miR-BART-7 at that time a well-recognized condition of
miR-BART-15 miR-BART-8 unknown aetiology. The link between EBV
miR-BART-5 miR-BART-9
miR-BART-16 miR-BART-22 and IM was confirmed in a study of serum
miR-BART-17 miR-BART-10 from Yale University students before and
miR-BART-6 miR-BART-11 after a diagnosis of IM78. The detection of
miR-BART-12
miR-BART-19 EBV in saliva from IM patients suggested
miR-BART-20 that oral transfer might be the route of
miR-BART-13
miR-BART-14 transmission79. In 1985, EBV replication
was observed in oral ‘hairy’ leukoplakia —
Figure 2 | EBV and its latent genes in 2016. a | Location and transcription of Epstein–Barr virus (EBV) white plaques on the tongue and oral tissues
Nature Reviews | Cancer
latent genes on the double-stranded viral DNA episome updated from the original figure shown in an commonly seen in male homosexuals with
Oncogene article in 2003 (REF. 265). The origin of plasmid replication (OriP) is shown in orange. The
human immunodeficiency virus (HIV)
short thick green arrows represent exons encoding latent proteins: six Epstein–Barr nuclear antigens
(EBNAs 1, 2, 3A, 3B and 3C, and EBNA‑LP), latent membrane proteins (LMPs 1, 2A and 2B), BamHI — suggesting that primary EBV infection
fragment H rightward open reading frame (BHRF1) and BamHI-A fragment rightward reading frame might involve virus replication in oral
(BARF1). The short blue arrows at the top represent the highly transcribed non-polyadenylated EBV- epithelial cells; however, it is still uncertain
encoded RNAs (EBER1 and EBER2). The middle long green arrow represents EBV transcription during whether this is the case80. Recent studies in
latency III, in which all the EBNAs are transcribed from either the Cp or Wp promoter; the different an organotypic keratinocyte culture system
EBNAs are encoded by individual mRNAs that are generated by differential splicing of the same long support a role for epithelial cells in the
primary transcript. The inner red arrow represents the EBNA1 transcript, which originates from the Qp replication and spread of EBV39.
promoter during latency I and latency II. Latency II is characterized by expression of EBNA1 together In 1998, David Thorley-Lawson81 showed
with the LMPs. Wp-restricted latency is initiated from the Wp promoter and there is expression of all that EBV-positive cells in the blood of
the EBNAs, except EBNA2, which is deleted in this form of latency (outer long blue arrow). b | Location
asymptomatic carriers are memory B cells.
of open reading frames for EBV latent proteins on the BamHI restriction map of the prototype EBV
B95‑8 genome and including the viral microRNAs (miRs) in sequence order. The BamHI fragments are How the virus gets into memory B cells
named according to size, with A being the largest. Lower-case letters indicate the smallest fragments. remains controversial. One model proposes
TR refers to the terminal repeats at each end of the genome. This region was often referred to as Nhet that memory B cells are directly infected
to indicate heterogeneity in this region according to the number of TRs within different virus isolates. with EBV82,83 (FIG. 3). Another, the germinal
BART, BamHI‑A rightward transcript. Adapted from REF. 265, Nature Publishing Group. centre model, proposes that EBV initially

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

infects naive B cells and expresses its growth surveillance in preventing lymphoma with intense immunosuppression, similar
programme (latency III), which drives B cell development was fully appreciated, when tumours, not all of which are EBV positive,
proliferation84. EBV-infected B cells then several reports of lymphoma arising in renal occur several years or more after solid organ
enter a germinal centre reaction and express allograft recipients immunosuppressed to transplantation in patients on low-level
the default programme (latency II); LMP1 prevent graft rejection were published104,105. immunosuppression for life112. Collectively,
and LMP2, as mimics of CD40 (also known The first hints that EBV might the term post-transplant lymphoproliferative
as TNFRSF5) and B cell receptor (BCR) be involved in the development of disease (PTLD) has been used to describe
signalling, respectively, probably mediate post-transplant lymphomas came from this heterogeneous group of conditions.
the survival of EBV-infected B cells in the a report in 1980 of raised antibody titres Many cases of early-onset PTLD are
germinal centre and their subsequent exit to EBV viral capsid antigen (VCA) oligoclonal and are regarded as outgrowths
as memory cells84–93. EBV-infected memory in transplant patients treated with of EBV-infected B cells, whereas others
B cells downregulate virus gene expression cyclosporin A106. Later that same year, resemble monoclonal lymphomas,
(latency 0) to avoid immune recognition. Dorothy Crawford107 identified EBNA commonly diffuse large B cell lymphoma
EBNA1 is expressed (latency I) during the protein in post-transplant lymphoma tissues. (DLBCL)110. Most PTLDs display an
proliferation of memory B cells. Activation This report was quickly followed by others unrestricted pattern of virus latency in which
of EBV-infected memory B cells can lead documenting EBV in lymphomas arising in EBV’s transforming proteins, EBNA2 and
to their differentiation into plasma cells, a other solid organ transplant recipients, and LMP1, are expressed. The EBNA3 family
process that has been shown to switch on the in bone marrow transplant patients108–110. members, which are highly immunogenic,
EBV lytic cycle with subsequent production Most lymphomas in bone marrow transplant are also expressed. In these cases the
of new virions94 (FIG. 3). recipients are of donor origin111. As well as virus growth transformation programme
The identification that humans evoke these early-onset forms of post-transplant proceeds unchecked113. However, many
humoral responses to EBV proteins provided lymphoma, occurring within weeks or of these tumours remain sensitive to
some of the key evidence linking the virus to months after transplantation and associated control by EBV-specific T lymphocytes114.
tumour development 40–44,78. However, in the
period after the initial discovery of EBV, little
was known about the nature of cell-mediated Box 1 | Strain variation
immunity to EBV. After it was shown Publication of the complete B95‑8 Epstein–Barr virus (EBV) sequence in 1984 represented the
that the high lymphocyte count that was largest DNA sequence to be determined16. In 2003, the wild-type EBV sequence was constructed
characteristic of IM was dominated mainly using B95‑8 as a backbone and Raji-derived sequences to replace the 12‑kb deletion in B95‑8
by T lymphocytes and not, as expected, (REF. 19). The current reference EBV sequence was released in 2010 (GenBank accession number
transformed B cells95,96, Denis Moss and NC_007605).
Rickinson set about unravelling the possible There are two EBV types, type 1 and type 2, which differ mainly in the sequences of the
contribution of T cells to EBV-specific Epstein–Barr nuclear antigen 2 (EBNA2) and EBNA3 genes17,18. Type 1 strains are more prevalent
immunity. They showed that in vitro worldwide and have greater transforming potential210,211.
Although there is a high level of similarity between EBV strains, variations exist in some viral genes,
infection of blood lymphocytes with EBV
mainly in the latent genes, that give rise to functional differences.
led to an initial phase of B cell proliferation, Complete genomes of EBV strains published to date are shown in the table.
but after a week or two the EBV-infected
B cells died — a phenomenon they termed EBV strain Type Origin Disease Year Refs
regression. They showed that regression was
B95‑8/Raji 1 USA and Nigeria IM and BL 2003 19
mediated by EBV-specific T cells present in
the blood of all healthy EBV-seropositive GD1 1 China NPC 2005 20
individuals97,98, and they were able to isolate AG876 2 Ghana BL 2006 21
CD4+ and CD8+ cytotoxic T cell (CTL) GD2 1 China NPC 2011 22
clones that recognized autologous LCLs99.
Using these clones to screen a series of HKNPC1 1 Hong Kong NPC 2012 23
peptides, they identified the first EBV CTL Akata 1 Japan BL 2013 24
epitope in 1990 (REF. 100), heralding a new Mutu 1 Kenya BL 2013 24
era in our understanding of the nature of M81 1 Hong Kong NPC 2013 25
T cell-mediated responses to EBV proteins
during virus infection, as well as stimulating K4123‑Mi and 1 USA LCLs 2013 26
K4413‑Mi
novel approaches to the rational design of
both prophylactic and therapeutic vaccines HKNPC2–HKNPC9 1 Hong Kong NPC 2014 27
against EBV101–103. NA19114, NA19315 1 USA LCLs 2014 28
and NA19384
EBV and other lymphomas 71 EBV strains 60 type 1 UK, Germany, Healthy 2015 29
Post-transplant lymphoproliferative and Hong Kong, China, Japan, saliva, LCLs,
disease. Although it was known that patients 11 type 2 S. Korea, Australia, Kenya PTLD, HL,
and Nigeria BL, NPC and
with various deficiencies of humoral or EBV-GC
cell-mediated immunity had an increased
BL, Burkitt lymphoma; EBV-GC, EBV-associated gastric carcinoma; HL, Hodgkin lymphoma; IM, infectious
lymphoma risk, it was not until the late mononucleosis; LCLs, lymphoblastoid cell lines; NPC, nasopharyngeal carcinoma; PTLD, post-transplant
1960s that the importance of immune lymphoproliferative disease.

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

Box 2 | Function of EBV latent genes LMPs as adjuvant therapy remained in


remission for a median of 3.1 years following
Latent proteins CTL infusion; of 21 patients with relapsed or
• Epstein–Barr nuclear antigen 1 (EBNA1) resistant disease at the time of CTL infusion,
-- Essential for episomal maintenance and replication212 13 had clinical responses, including 11
-- Induces B cell lymphomas in transgenic mice213 complete responses119.
-- Enhances cell survival68 It should be noted that EBV-positive
-- Induces genetic instability214 DLBCL also arises in patients, including
• EBNA2 children, without any previous history of
-- Discovered when a laboratory-derived Epstein–Barr virus (EBV) strain, P3HR‑1, with an EBNA2 immunosuppression; in older patients it
deletion was unable to transform B cells in vitro215
is assumed that a decline in EBV-specific
-- Mimics a constitutively activated NOTCH receptor216
immunity associated with ageing is
• EBNA3A and EBNA3C
responsible120.
-- Essential for in vitro B cell transformation217
-- Cooperate with oncogenic HRAS in transforming primary rodent fibroblasts218
X‑linked lymphoproliferative disease
• EBNA3B
type 1. David Purtilo121 provided the first
-- May function as a tumour suppressor in diffuse large B cell lymphoma (DLBCL)219
detailed characterization of ‘Duncan’s
• EBNA leader protein (EBNA‑LP)
syndrome’, later to be called X‑linked
-- Required for the efficient outgrowth of lymphoblastoid cell lines (LCLs)220
lympho­proliferative disease type 1 (XLP1).
• Latent membrane protein 1 (LMP1)
Boys with XLP1 develop a severe, often
-- Classic oncogene221
-- Essential for in vitro B cell transformation222
fatal, IM within weeks after primary EBV
-- Functions as a constitutively activated tumour necrosis factor receptor (TNFR)145 and resembles infection. This may be accompanied by
CD40 receptor in vivo85 a condition known as haemophagocytic
-- Activates signalling pathways that contribute to its oncogenic effects223 lymphohistiocytosis, in which there is
• LMP2A and LMP2B uncontrolled proliferation of activated
-- Identified in 1990 (REF. 224) lymphocytes and macrophages that secrete
-- LMP2A is a B cell receptor (BCR) mimic that can rescue BCR-deficient B cells from apoptosis86 high levels of inflammatory cytokines.
• BamHI fragment H rightward open reading frame 1 (BHRF1) Survivors have multiple immune defects,
-- BCL‑2 homologue originally thought to be a lytic gene225,226 including reduced levels of antibodies
-- Important for apoptotic protection in Wp‑restricted Burkitt lymphoma227 in the blood and a propensity to develop
• BamHI‑A fragment rightward reading frame 1 (BARF1) lymphoma121,122. In 1998, an international
-- Induces transformation of rodent fibroblasts and a human B cell line228,229 consortium of scientists published three
-- Reported as an early lytic gene230 papers describing the genetic defect in
-- Expressed in nasopharyngeal carcinoma (NPC) and EBV-gastric cancer in the absence of lytic XLP1 as a mutation in the SH2 domain
gene expression231 containing 1A (SH2D1A) gene, which
Latent non-coding RNA encodes a defective signalling lymphocyte
EBV was the first human virus found to encode microRNAs (mi­RNAs)232. activation molecule (SLAM)-associated
• BamHI‑A rightward transcript mi­RNAs (BART mi­RNAs) protein (SAP)123–125. Thus, patients with
-- 44 mature BART mi­RNAs conclusively verified to date233 XLP1 have impaired T cell and natural killer
-- Maintain latency by targeting EBV lytic genes, modulating LMP1 expression, targeting (NK) cell engagement of B cells rather than
pro-apoptotic proteins, impairing host immune responses and inactivating tumour a specific inability to cope with EBV itself 126.
suppressor genes234–240
-- Can drive tumour growth in vivo241,242
Hodgkin lymphoma: another addition to
-- Potential serum biomarkers in patients with NPC201,243
the EBV-positive B cell lymphoma family.
• BHRF1 mi­RNAs
Hodgkin lymphoma (HL) is an unusual
-- Three BHRF1 mi­RNAs are encoded232
-- Important for B cell transformation244 tumour characterized by the presence of
rare, large, often binucleated, tumour cells,
• EBV-encoded RNA 1 (EBER1) and EBER2
-- Are highly abundant non-coding RNAs245
known as Hodgkin–Reed–Sternberg (HRS)
-- Form stem–loop structures giving rise to double-stranded RNA-like molecules246 cells, which are surrounded by a prominent
-- Interact with a number of proteins and contribute to the activation of innate immunity247 microenvironment composed of various
non-malignant inflammatory cells, including
T cells, macrophages and fibroblasts.
Early therapies for PTLD relied on surgery to EBV has led to the development of HL displays a bimodal age distribution in
with irradiation, chemotherapy and, in adoptive CTL therapy for PTLD and other developed countries: the first peak occurs
some cases, treatment with antiviral drugs, EBV-related tumours. This approach, between the ages of 15 and 34 years, and the
but mortality was high115. Thomas Starzl pioneered by Cliona Rooney, is especially second at older than 50 years127. In contrast,
and colleagues116 were the first to show that successful against EBV-positive lymphomas in developing countries, the young adult
reduction or discontinuance of immuno- expressing the latency III programme117,118. peak is less conspicuous and childhood
suppression caused regression of the lesions, For example, in a recent clinical trial, 28 of rates are higher 128. It has been suggested
often without subsequent rejection of the 29 high-risk or multiple-relapse patients that the increased HL risk in young adults
grafts. A better understanding of immunity with lymphoma receiving CTLs against in developed countries might be due to

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

late exposure to a common infectious Genetic studies also show that different this stage of HIV infection152,153. Thus, like
agent. In 1974, two studies suggested that alleles of the human leukocyte antigen A malaria, HIV probably acts as a chronic
EBV might be this infectious agent; both (HLA‑A) gene are associated with different B cell stimulator that increases the pool of
reported a significantly increased risk of susceptibilities to both EBV-positive HL and EBV-infected B cells carrying accidental
HL in individuals with a previous history IM. Most notably, individuals possessing the MYC translocations. The incidence of HL
of IM129,130. In 1985, Sibrand Poppema131 HLA‑A*01 variant have an increased risk of in patients with HIV also increases as CD4+
provided the first direct evidence of EBV’s both EBV-positive HL and IM, and those T cell counts begin to fall but reaches a
involvement when he detected EBNA with the HLA‑A*02 variant have a decreased peak with intermediate levels of immune
protein in the HRS cells of a single patient. risk of both conditions147,148. Because HLA‑A impairment and declines thereafter in
EBV DNA was then detected in whole proteins are critical for antigen presentation severely immunocompromised patients154,
HL biopsies and later in HRS cells132,133. to CTLs, these observations point to the suggesting that CD4+ T cells contribute to
In 1990, Richard Ambinder 134 used in situ importance of immune responses to EBV HL pathogenesis.
hybridization to detect expression of the as potentially shared pathogenic triggers in
EBERs in HRS cells; this is now the gold EBV-positive HL and IM147,148. More surprises: EBV-associated diseases
standard approach for latent EBV detection of NK cells and T cells. Although it was
in pathology samples. HIV and EBV-associated B cell lymphomas. originally thought that EBV could only
Despite the strong linkage to EBV, the Early in the 1980s, reports appeared of infect lymphocytes of the B cell lineage,
B cell origin of HL was demonstrated only the increased incidence of lymphomas in in 1988, James Jones and colleagues155
in 1994 when Ralf Küppers showed that HRS patients with the newly described acquired found EBV genomes in the CD4+ T cells
cells harbouring clonal immunoglobulin immunodeficiency syndrome (AIDS). The of three patients with T cell lymphoma.
rearrangements are somatically first report was of a case in a homosexual Subsequently, EBV was found in the CD4+
hypermutated, and in some cases have man, who had raised antibody titres to T cells of a boy with chronic active EBV
mutations that destroy the coding capacity of VCA149. Soon after, John Ziegler 150 reported infection (CAEBV), a life-­threatening
originally functional immunoglobulin genes four cases of BL in homosexual men; of the condition characterized by severe persistent
(crippling mutations)135. Thus, HRS cells are three tumours with available tissue, two or progressive IM156. Two years later,
thought to be derived from germinal centre were EBNA positive as determined by ACIF. extranodal natural killer (ENK)/T cell
B cells that lack a functional BCR and that Thereafter, it was shown that BL in AIDS lymphoma, an aggressive form of lymphoma
must have escaped apoptosis, which would carries the typical MYC translocations151. that expresses some T cell markers and is
otherwise have been their normal fate. Unlike DLBCL, which is also increased in usually of NK cell origin, was also found
Two pieces of evidence suggest that EBV HIV infection but only when the patient is to be consistently EBV positive157. We
can provide this anti-apoptotic function: severely immunocompromised, BL occurs now know that CAEBV, haemophagocytic
crippling immunoglobulin mutations are early in HIV infection when circulating lympho­histiocytosis (discussed above)
found almost exclusively in EBV-positive CD4+ T cell numbers are relatively normal; and ENK/T cell lymphoma represent a
patients136, and EBV can efficiently this is probably the result of expanded spectrum of disease often characterized by
immortalize BCR-negative germinal centre germinal centre activity characteristic of EBV infection of T or NK cells; CAEBV
B cells in vitro137–139. LMP1 and LMP2A,
which are expressed in EBV-positive
HRS cells, are thought to provide these Box 3 | Functions of EBV lytic genes
anti-apoptotic signals by mimicking
CD40 receptor and BCR functions, Two Epstein–Barr virus (EBV) genes, BZLF1 and BRLF1, synergistically induce the expression
of multiple early lytic genes, including those that encode the six viral proteins required to
respectively 140–145. Thus, the mechanism
assemble the replication machinery; BALF5 (the DNA polymerase), BALF2 (the single-stranded
of protection in HL is distinct from that DNA-binding protein homologue), BMRF1 (the DNA polymerase processivity factor), BSLF1
observed in BL, probably reflecting the (the primase homologue), BBLF4 (the helicase homologue) and BBLF2/3 (formed from the splicing
different apoptotic stimuli that must be of BBLF2 and BBLF3 open reading frames; a potential homologue of the third component of the
overcome in each tumour. helicase–primase complex)248. In addition to its function as a transactivator, direct interactions of
Although the epidemiology had initially the BZLF1 protein with the lytic origin of DNA replication (oriLyt), and the viral replication
provided the impetus for the search for proteins, were found to be essential for EBV DNA replication249–251. In 2007, it was shown that lytic
EBV in HL, contrary to expectations it was cycle transcription occurs following primary infection of B cells252, a phenomenon that was
shown that in developed countries, EBV subsequently confirmed in epithelial cells253. However, a full lytic cycle immediately post-infection
is present mostly in a histological variant cannot be induced because BZLF1 does not bind to a viral genome that is devoid of CpG
methylation254. Thus, an abortive lytic cycle has been proposed to be the initial mode of primary
of HL known as mixed cellularity HL that
EBV infection that is considered essential for the transformation of resting B cells255. Several lytic
occurs mainly in children and older adults, genes encode immune evasion proteins, including BNLF2a (inhibits the transporter of antigen
and is found less frequently in the other processing)256, BILF1 (induces major histocompatibility complex (MHC) class I internalization and
major histological variant, known as nodular degradation)257 and BGLF5 (mediates host shut-off)258.
sclerosis HL, which makes up most of the Evidence for a contribution of the lytic cycle to EBV-induced oncogenesis has emerged only in
young adult peak. Nevertheless, a small recent years. For example, the presence of low numbers of lytically infected cells could enhance
fraction of young adult HLs are EBV positive tumour growth through the release of growth factors and immunosuppressive cytokines259,260.
and some are preceded by IM; we now know EBV lytic genes BZLF1, BGLF4 and BGLF5 can also induce genome instability261–263. Recent RNA
that a prior diagnosis of IM is associated sequencing and high-throughput PCR array analyses have revealed high-level expression of lytic
with an increased risk of developing genes, particularly LF1, LF2, LF3, BILF1, BALF4 and BHLF1, in cell lines and tumour biopsies,
although it is not clear whether these genes are expressed in latently infected cells77,264.
EBV-positive, but not EBV-negative, HL146,147.

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

often develops into either ENK/T cell contributes to the excessive production of lytic cycle proteins163. Further serological
lymphoma155 or haemophagocytic lympho­ pro-­inflammatory cytokines characteristic analysis showed an association between
histiocytosis158. ENK/T cell lymphomas of ENK/T cell lymphomas162. EBV antibody titres and NPC tumour
are more common in Asian populations159 stage164, and identified VCA-specific
and express a virus latency II pattern with EBV and epithelial cancers immunoglobulin A (IgA) levels as a
variable LMP1 expression160. Recent work NPC. A surprising early observation made potential prognostic indicator 165. The
demonstrates that EBV type 2 strains in 1966 was that the BL antigen-specific apparent specificity of the association
(BOX 1) preferentially infect primary T cells antibodies described by Old et al.40 were between VCA IgA levels and NPC led
in vitro, resulting in expression of latent also detectable in the serum of patients Zeng Yi to embark on mass serological
genes and leading to T cell activation and with “post-nasal space carcinomas” or, screening programmes in Wuzhou City,
proliferation, with alteration of cytokine as it later became known, NPC. Patients China. These studies not only confirmed
profiles161. These preliminary observations with NPC were subsequently shown to that IgA responses to VCA and early
are consistent with data showing that LMP1 have elevated titres of antibodies to several antigen were detectable in patients with
NPC, but also revealed that they were
present in some patients with hitherto
Germinal centre model
unrecognized NPC166. These data suggest
EBV that EBV-based screening programmes
Germinal centre could reduce mortality from NPC through
Plasma cell EBV replication
early detection and treatment. The approach
to screening for early NPC has been greatly
enhanced recently by the use of quantitative
Naive Latency II
B cell PCR to detect circulating EBV DNA167.
(EBNA1, LMP1 and LMP2)
Latency III In fact, circulating cell-free EBV DNA has
(all EBNAs and LMPs) become a gold standard biomarker that
Latency 0 Latency I
(no protein expression) (EBNA1) can be used to stage patients and provide
prognostic information168.
Direct infection model Although zur Hausen55 had already
shown that extracts of NPC biopsies
contained EBV DNA, crucially, in 1973, EBV
? DNA was detected by in situ hybridization in
NPC tumour cells and not in the infiltrating
Memory lymphoid cells as had been expected169.
B cell Normal persistence Subsequent studies showed that NPC
? ?
Latency III tumour cells carry clonal EBV genomes170
B cell lymphomas and express a virus latency II pattern
with variable LMP1 expression32,171–173.
It is the most undifferentiated type of NPC
Burkitt lymphoma Hodgkin lymphoma PTLD and DLBCL (so‑called non-keratinizing type II and
Latency I Latency II Latency III type III tumours) that are predominantly
EBV positive174. Tumours histologically
Figure 3 | Virus persistence in the B cells of the human host and the origin of EBV-associated similar to undifferentiated NPC can be
Nature Reviews | Cancer
B cell lymphomas. Upper panel: normal persistence. Epstein–Barr virus (EBV) resides in found at other sites, such as the stomach
memory B cells of asymptomatic hosts. There are two models to explain how the virus enters mem‑ and salivary glands, and are also frequently
ory B cells. In the germinal centre model, EBV infection of naive B cells leads to a latency III growth EBV positive175.
programme, in which the proliferation and expansion of the infected B cell pool is driven by expres‑ A scheme for NPC pathogenesis has been
sion of all EBV latent genes. Cells then enter the germinal centre and express latency II (default
proposed whereby loss of heterozygosity
programme), characterized by expression of Epstein–Barr nuclear antigen 1 (EBNA1), latent mem‑
brane protein 1 (LMP1) and LMP2. LMP1 provides a CD40‑like signal and LMP2 a surrogate B cell (LOH) at chromosomes 3p and 9p occurs
receptor-like signal — mimicking the same signals as those provided to the antigen-specific as an early event 176, possibly as a result of
B cell in the normal germinal centre. EBV-infected germinal centre B cells leave and enter the mem‑ exposure to environmental cofactors such
ory B cell pool. Here, EBV protein expression is silenced (latency 0); these cells are replenished by as dietary components (that is, carcinogenic
the same signals that induce the proliferation of normal memory B cells. Proliferating EBV-infected volatile nitrosamines in salted fish or other
memory B cells require EBNA1 expression (latency I) for viral episome segregation. Memory B cells preserved food) and smoking 177, creating
can terminally differentiate into plasma cells (solid arrow), which triggers virus replication. EBV- low-grade pre-invasive lesions that, after
infected germinal centre B cells might also differentiate directly into plasma cells (dashed arrow). additional genetic and epigenetic events,
In the direct infection model (shown below), EBV accesses memory B cells following the direct become susceptible to EBV infection4,178
infection of these cells, which may involve a latency III intermediary. Lower panel: the origin of the
(FIG. 4). Once infected, EBV latent genes
EBV-associated B cell lymphomas. There is uncertainty about the exact stages of differentiation
from which the EBV-positive B cell lymphomas arise (as indicated by the black dotted lines), as it provide growth and survival benefits,
cannot be assumed that the pattern of latency observed in the progenitor cell is recapitulated in resulting in the development of NPC.
the corresponding tumour. The figure illustrates the presumed (question marks indicate this uncer‑ Additional genetic and epigenetic changes
tainty) cell of origin based on current evidence for post-transplant lymphoproliferative disease occur after EBV infection. Detailed analysis
(PTLD), diffuse large B cell lymphoma (DLBCL), Hodgkin lymphoma and Burkitt lymphoma. of the genomic landscape of NPC has

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

Normal epithelium Pre-invasive lesions NPC


CIS?

Low grade High grade

Epithelium

Superficial zone

Midzone

Basal layer
Basement
membrane Dermis

LOH on chromosomes 3p and 9p

Overexpression of CCND1 and BCL-2; increased telomerase activity

Inactivation of RASSF1A, RARB2, CDKN2A and DAPK1

EBV latent infection

LOH on chromosomes 11q, 13q, 14q and 16q

Other genetic changes (for example inactivation of EDNRB)

Figure 4 | Schematic representation of the pathogenesis of NPC. cofactors such as dietary components (for example, salted fish), creating
In nasopharyngeal carcinoma (NPC), the tumour cells carry monoclonal low-grade pre-invasive lesions that after additional genetic and epi­
viral genomes indicating that Epstein–Barr virus (EBV) infection must have genetic events become susceptible to stable Nature Reviews | Cancer
EBV infection. Once
occurred before expansion of the malignant cell clone170. EBV infection is infected, EBV latent genes provide growth and survival benefits that result
detected in high-grade pre-invasive lesions in NPC but not in low-grade in the development of NPC. Additional genetic (for example, LOH on
disease170. Multiple genetic changes have been found in NPC, with fre‑ chromosomes 11q, 13q, 14q and 16q) and epigenetic changes occur after
quent chromosomal deletions and promoter hypermethylation of specific EBV infection and contribute to metastatic disease176,270. The precise tim‑
genes on chromosomes 3p, 9p and 11q266. Deletions in both 3p and 9p ing of epigenetic events in the pathway of NPC development is unknown,
have been identified in low-grade EBV-negative dysplastic lesions and in although some changes such as RAS association domain family member 1
normal nasopharyngeal epithelium from individuals who are at high risk (RASSF1A) inactivation may occur early, whereas others (for example,
of developing NPC. In vitro studies demonstrate that cyclin D1 (CCND1) inactivation of retinoic acid receptor β2 (RARB2), CDKN2A and death-­
overexpression (a consequence of cyclin-dependent kinase inhibitor 2A associated protein kinase 1 (DAPK1)) may be influenced by the ability of
(CDKN2A, which encodes p16INK4A) deletion on chromosome 9p and ampli‑ EBV to enhance the activity of the methylation machinery271. Recent
fication of the CCND1 locus on chromosome 11q) facilitates persistent analy­sis of the genomic landscape in NPC has confirmed a role for chro‑
EBV infection of immortalized nasopharyngeal epithelial cells 267. matin modification in the carcinogenic process and identified additional
Increased expression of the anti-apoptotic protein BCL‑2 (REF. 268) and somatic events, including mutations in the ERBB–PI3K signalling path‑
elevated telomerase activity269 in low-grade dysplastic lesions may also way179. The pathogenesis of EBV-associated gastric cancer is similar to
promote the establishment of latent EBV infection. A scheme has been that of NPC, with premalignant lesions occurring before EBV infection190
proposed whereby loss of heterozygosity (LOH) occurs early in the patho‑ and a distinctive hypermethylation phenotype188. CIS, carcinoma in situ;
genesis of NPC, possibly as a result of exposure to environmental EDNRB, endothelin receptor B.

revealed particular pathways (for example, confirmed that approximately 10% of that may be attributed to ethnic and genetic
chromatin modification and ERBB–PI3K these tumours are EBV positive4. Given differences182,183. EBV-GCs have distinct
signalling) that may work in concert the worldwide prevalence of gastric cancer phenotypic and clinical characteristics
with EBV to drive the development of (GC) (952,000 cases in 2012)181, EBV‑GC compared with EBV-negative GC, including
this tumour 179. may represent the most common form of loss of cyclin-dependent kinase (CDK)
EBV-associated malignancy 182,183. EBV‑GC inhibitor p16INK4A (encoded by CDKN2A)
Gastric cancer. In 1992, EBV was detected displays a viral latency pattern similar to that expression, high CpG island methylator
in the tumour cells of typical gastric of NPC33. There is substantial geographical phenotype (CIMP), the presence of
adenocarcinoma180. Subsequent studies variation in the association of EBV with GC wild-type p53, apparent driver mutations in

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

the PI3K catalytic subunit-α (PIK3CA) and epithelial cells in this process. The impact EBV is the ultimate biomarker, and routine
AT‑rich interactive domain 1A (ARID1A) of EBV infection on the development of application of serum testing for virus
genes, and better patient survival184–189. NPC and EBV‑GC may be a consequence DNA could prove useful in detecting
As in NPC, the precise role of EBV in of the aberrant establishment of virus early disease, providing prognostic
the pathogenesis of GC is unknown, but the latency in epithelial cells that have already information, and in monitoring patients
absence of EBV infection in premalignant undergone premalignant changes — a after treatment, approaches that may be
gastric lesions supports the contention that pathogenic process that is distinct from further enhanced by the development of
virus infection is a relatively late event190. EBV-induced lymphomagenesis in which additional adjunctive tests (for example, to
Epidemiological studies show that EBV‑GC the virus appears to be the initiator. Unlike detect EBV mi­RNAs and methylated tumour
is related to birth order (younger siblings receptor-mediated infection of B cells, suppressor genes)201,202. The development
being more susceptible possibly as a result of cell‑to‑cell infection is well recognized as of novel therapeutic approaches using virus
earlier exposure to EBV), high salt intake and the main pathway for EBV infection in reactivation203, gene therapy 204, drugs that
exposure to metal dust, but these factors may epithelial cells195,196. Recently, a novel in‑cell target EBV latent protein function, such as
vary geographically (for example, between infection mechanism was described for inhibitors of EBNA1, which is known to be
Japan and Colombia, which both have EBV infection of nasopharyngeal epithelial expressed in every EBV-positive tumour 205,206,
high rates of GC), supporting the need for cells through the formation of heterotypic or therapeutic vaccination207,208 provides
more detailed investigation182,183. It has been cell‑in‑cell structures (invasion of B cells exciting opportunities for meaningful clinical
suggested that distinct EBV strains contribute into epithelial cells by an as yet unknown intervention. The effectiveness of these
to the development of EBV‑GC, although this mechanism), a process that might be modalities will be further enhanced by the
could simply reflect geographical variation related to the maintenance of EBV latency molecular classification and stratification
whereby EBV strains common in the general in epithelial cells197. The development of of cancer patients with EBV-associated
population of certain countries are more able more efficient in vitro systems for studying tumours. Although there is renewed
to promote GC development191. EBV infection and replication in different interest in the possibility of a prophylactic
cell types is helping to unravel the complex vaccine against EBV101, there are substantial
Future perspectives interplay between the virus and the host cell. economic and practical hurdles in the
EBV was discovered more than 50 years ago The use of recombinant EBVs continues to implementation of this approach, along
and remains the most common persistent shed light on the role of viral genes in the with concerns about the possibility of
asymptomatic virus infection in humans. transformation process, on the requirements unintended consequences of eradication
The life-long relationship between EBV and for the efficient production of progeny virus of EBV infection; for example, one study
its immune host is a consequence of the and on the role of EBV-encoded membrane has indicated that herpesvirus latency
ability of the virus to persist in the memory glycoproteins in the infection process. confers symbiotic protection from bacterial
B cell pool of normal healthy individuals. The key question of the role of EBV strain infection209. Our growing understanding
Such an intimate host–pathogen interaction variation in oncogenesis is becoming more of EBV-associated oncogenesis provides
is reflected by the presence of EBV-like tractable with the advent of more sensitive paradigms for the development of targeted
B cell trophic viruses (lymphocrypto­ virus genome sequencing technologies29 cancer therapies and diagnostics and further
viruses) in non-human primates, suggesting and the ability to clone wild-type EBV confirms the far-reaching value of tumour
that EBV has co‑evolved with its host 192. strains for biological charac­terization25,198. virology to the whole cancer field.
The ability of EBV to transform B cells Understanding the host cell–virus interaction L.S.Y. is at Warwick Medical School, The University of
in vitro has contributed to the unequivocal will depend on the generation of appropriate Warwick, Coventry CV4 7AL, UK.
identification of EBV as oncogenic in in vitro and in vivo models, particularly those L.F.Y. is at the Department of Oral and Craniofacial
humans and to a deeper understanding that allow an understanding of the role of the Sciences and Oral Cancer Research Coordinating
of the role of the virus in B cell tumours. tumour microenvironment, including the Centre, Faculty of Dentistry, University of Malaya,
The precise role of EBV in the pathogenesis local cytokine milieu. Kuala Lumpur, Malaysia.
of epithelial cancers, including NPC and A characteristic feature of many P.G.M. is at the Institute of Cancer and Genomic
EBV‑GC, remains unclear. The detection EBV-associated cancers is the involvement Medicine, University of Birmingham, Vincent Drive,
Edgbaston, Birmingham B15 2TT, UK.
of EBV in other rarer forms of cancer, of chronic inflammation. This is exemplified
such as leiomyosarcomas in immuno­ by tumours such as BL and is also apparent Correspondence to L.S.Y.
suppressed children, not only underlines the in other rarer forms of EBV-positive cancer L.S.Young@warwick.ac.uk

importance of disturbed immunity in viral arising at sites of long-term persistent doi:10.1038/nrc.2016.92


Published online 30 September 2016
oncogenesis but also provides a striking inflammation, such as pyothorax-
example of the harmful consequences and chronic osteomyelitis-associated 1. De Martel, C. et al. Global burden of cancers
attributable to infections in 2008: a review and
that can follow the aberrant infection of lymphomas199, in so-called ‘remnant’ GCs synthetic analysis. Lancet Oncol. 13, 607–615
other cell types193. The complex interplay that develop after partial gastrectomy 190 (2012).
2. Khan, G. & Hashim, M. J. Global burden of deaths
between EBV and the immune system can and in certain benign conditions such as from Epstein–Barr virus: attributable malignancies
also manifest itself in other unexpected muco-cutaneous ulcers200. Unravelling 1990–2010. Infect. Agent. Cancer 9, 38 (2014).
3. Crawford, D. H., Rickinson, A. B. & Johannessen, I. in
ways, as demonstrated by increasing the interplay between virus infection and Cancer Virus: The Story of Epstein–Barr Virus (Oxford
evidence causally implicating EBV in the inflammation will be an important priority Univ. Press, 2014).
4. Tsao, S. W., Tsang, C. M., To, K. F. & Lo, K. W. The role
pathogenesis of multiple sclerosis194. for future studies. of Epstein–Barr virus in epithelial malignancies.
Little is known about the replicative Whatever the role of EBV in the J. Pathol. 235, 323–333 (2015).
5. Pasteur, L. Inaugural address as newly appointed
life cycle of EBV in vivo particularly with oncogenic process, there is an opportunity Professor and Dean at the opening of the new Faculte
regard to the relative role of B cells versus to exploit this association for patient benefit. des Sciences, Univ. Lille (7 Dec 1854).

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

6. Epstein, M. A. On the discovery of Epstein–Barr 31. Rowe, M., Lear, A. L., Croom-Carter, D., Davies, A. H. 55. zur Hausen, H. et al. EBV DNA in biopsies of Burkitt
virus: a memoir. Epstein–Barr Virus Rep. 6, 58–63 & Rickinson, A. B. Three pathways of Epstein–Barr tumours and anaplastic carcinomas of the
(1999). virus gene activation from EBNA1‑positive latency in nasopharynx. Nature 228, 1056–1058 (1970).
7. Epstein, M. A., Achong, B. G. & Barr, Y. M. Virus B lymphocytes. J. Virol. 66, 122–131 (1992). 56. Reedman, B. M. et al. Epstein–Barr virus-associated
particles in cultured lymphoblasts from Burkitt’s 32. Young, L. S. et al. Epstein–Barr virus gene expression complement-fixing and nuclear antigens in Burkitt
lymphoma. Lancet 1, 702–703 (1964). in nasopharyngeal carcinoma. J. Gen. Virol. 69, lymphoma biopsies. Int. J. Cancer 13, 755–763
8. Epstein, M. A. Citation classic – Virus particles in 1051–1065 (1988). (1974).
cultured lymphoblasts from Burkitt’s lymphoma. Curr. 33. Imai, S. et al. Gastric carcinoma: monoclonal epithelial 57. Dillner, J. et al. Antibodies against a synthetic peptide
Contents Life Sci. 14, 156 (1979). malignant cells expressing Epstein–Barr virus latent identify the Epstein–Barr virus-determined nuclear
9. Pope, J. H., Achong, B. G., Epstein, M. A. & infection protein. Proc. Natl Acad. Sci. USA 91, antigen. Proc. Natl Acad. Sci. USA 81, 4652–4656
Biddulph, J. Burkitt lymphoma in New Guinea: 9131–9135 (1994). (1984).
establishment of a line of lymphoblasts in vitro and 34. zur Hausen, H., O’Neill, F. J., Freese, U. K. & Hecker, E. 58. Dillner, J., Kallin, B., Ehlin-Henriksson, B., Timar, L. &
description of their fine structure. J. Natl Cancer Inst. Persisting oncogenic herpesvirus induced by the Klein, G. Characterization of a second Epstein–Barr
39, 933–945 (1967). tumour promoter TPA. Nature 272, 373–375 (1978). virus-determined nuclear antigen associated with the
10. Henle, W., Diehl, V., Kohn, G., Zur Hausen, H. 35. Kallin, B., Luka, J. & Klein, G. Immunochemical BamHI WYH region of EBV DNA. Int. J. Cancer 35,
& Henle, G. Herpes-type virus and chromosome characterization of Epstein–Barr virus-associated 359–366 (1985).
marker in normal leukocytes after growth with early and late antigens in n‑butyrate-treated P3HR‑1 59. Manolov, G. & Manolova, Y. Marker band in one
irradiated Burkitt cells. Science 157, 1064–1065 cells. J. Virol. 32, 710–716 (1979). chromosome 14 from Burkitt lymphomas. Nature
(1967). 36. Countryman, J. & Miller, G. Activation of expression of 237, 33–34 (1972).
11. Pope, J. H. Establishment of cell lines from peripheral latent Epstein–Barr herpesvirus after gene transfer 60. Zech, L., Haglund, U., Nilsson, K. & Klein, G.
leucocytes in infectious mononucleosis. Nature 216, with a small cloned subfragment of heterogeneous Characteristic chromosomal abnormalities in biopsies
810–811 (1967). viral DNA. Proc. Natl Acad. Sci. USA 82, 4085–4089 and lymphoid-cell lines from patients with Burkitt and
12. Pope, J. H., Horne, M. K. & Scott, W. Transformation (1985). non-Burkitt lymphomas. Int. J. Cancer 17, 47–56
of foetal human keukocytes in vitro by filtrates of a 37. Bhende, P. M., Dickerson, S. J., Sun, X., Feng, W. H. & (1976).
human leukaemic cell line containing herpes-like virus. Kenney, S. C. X‑Box-binding protein 1 activates lytic 61. Bernheim, A., Berger, R. & Lenoir, G. [Translocations
Int. J. Cancer 3, 857–866 (1968). Epstein–Barr virus gene expression in combination t(2;8) and t(8;22) in continuous cell lines of African
13. Henderson, E., Miller, G., Robinson, J. & Heston, L. with protein kinase D. J. Virol. 81, 7363–7370 (2007). Burkitt’s lymphoma]. C. R. Seances Acad. Sci. D 291,
Efficiency of transformation of lymphocytes by 38. Sun, C. C. & Thorley-Lawson, D. A. Plasma cell-specific 237–239 (1980).
Epstein–Barr virus. Virology 76, 152–163 (1977). transcription factor XBP‑1s binds to and 62. Erikson, J., ar‑Rushdi, A., Drwinga, H. L.,
14. Sugden, B. & Mark, W. Clonal transformation of adult transactivates the Epstein–Barr virus BZLF1 Nowell, P. C. & Croce, C. M. Transcriptional activation
human leukocytes by Epstein–Barr virus. J. Virol. 23, promoter. J. Virol. 81, 13566–13577 (2007). of the translocated c‑myc oncogene in Burkitt
503–508 (1977). 39. Temple, R. M. et al. Efficient replication of Epstein– lymphoma. Proc. Natl Acad. Sci. USA 80, 820–824
15. Thorley-Lawson, D. A. & Allday, M. J. The curious case Barr virus in stratified epithelium in vitro. Proc. Natl (1983).
of the tumour virus: 50 years of Burkitt’s lymphoma. Acad. Sci. USA 111, 16544–16549 (2014). 63. Armelin, H. A. et al. Functional role for c‑myc in
Nat. Rev. Microbiol. 6, 913–924 (2008). 40. Old, L. J. et al. Precipitating antibody in human serum mitogenic response to platelet-derived growth factor.
16. Baer, R. et al. DNA sequence and expression of the to an antigen present in cultured Burkitt’s lymphoma Nature 310, 655–660 (1984).
B95‑8 Epstein–Barr virus genome. Nature 310, cells. Proc. Natl Acad. Sci. USA 56, 1699–1704 64. Einat, M., Resnitzky, D. & Kimchi, A. Close link
207–211 (1984). (1966). between reduction of c‑myc expression by interferon
17. Dambaugh, T., Hennessy, K., Chamnankit, L. & 41. Levy, J. A. & Henle, G. Indirect immunofluorescence and, G0/G1 arrest. Nature 313, 597–600 (1985).
Kieff, E. U2 region of Epstein–Barr virus DNA may tests with sera from African children and cultured 65. Evan, G. I. et al. Induction of apoptosis in fibroblasts
encode Epstein–Barr nuclear antigen 2. Proc. Natl Burkitt lymphoma cells. J. Bacteriol. 92, 275–276 by c‑myc protein. Cell 69, 119–128 (1992).
Acad. Sci. USA 81, 7632–7636 (1984). (1966). 66. Milner, A. E., Grand, R. J., Waters, C. M. &
18. Sample, J. et al. Epstein–Barr virus types 1 and 2 42. Henle, G. & Henle, W. Immunofluorescence in cells Gregory, C. D. Apoptosis in Burkitt lymphoma cells
differ in their EBNA‑3A, EBNA‑3B, and EBNA‑3C derived from Burkitt’s lymphoma. J. Bacteriol. 91, is driven by c‑myc. Oncogene 8, 3385–3391
genes. J. Virol. 64, 4084–4092 (1990). 1248–1256 (1966). (1993).
19. de Jesus, O. et al. Updated Epstein–Barr virus (EBV) 43. Henle, G. et al. Antibodies to Epstein–Barr virus in 67. Shimizu, N., Tanabe-Tochikura, A., Kuroiwa, Y. &
DNA sequence and analysis of a promoter for the Burkitt’s lymphoma and control groups. J. Natl Cancer Takada, K. Isolation of Epstein–Barr virus (EBV)-
BART (CST, BARF0) RNAs of EBV. J. Gen. Virol. 84, Inst. 43, 1147–1157 (1969). negative cell clones from the EBV-positive Burkitt’s
1443–1450 (2003). 44. de‑The, G. et al. Epidemiological evidence for causal lymphoma (BL) line Akata: malignant phenotypes
20. Zeng, M. S. et al. Genomic sequence analysis of relationship between Epstein–Barr virus and Burkitt’s of BL cells are dependent on EBV. J. Virol. 68,
Epstein–Barr virus strain GD1 from a nasopharyngeal lymphoma from Ugandan prospective study. Nature 6069–6073 (1994).
carcinoma patient. J. Virol. 79, 15323–15330 274, 756–761 (1978). 68. Kennedy, G., Komano, J. & Sugden, B. Epstein–Barr
(2005). 45. Whittle, H. C. et al. T‑cell control of Epstein–Barr virus- virus provides a survival factor to Burkitt’s lymphomas.
21. Dolan, A., Addison, C., Gatherer, D., Davison, A. J. & infected B cells is lost during P. falciparum malaria. Proc. Natl Acad. Sci. USA 100, 14269–14274
McGeoch, D. J. The genome of Epstein–Barr virus Nature 312, 449–450 (1984). (2003).
type 2 strain AG876. Virology 350, 164–170 46. Moormann, A. M. et al. Exposure to holoendemic 69. Ruf, I. K. et al. Epstein–Barr virus regulates c‑MYC,
(2006). malaria results in suppression of Epstein–Barr virus- apoptosis, and tumorigenicity in Burkitt lymphoma.
22. Liu, P. et al. Direct sequencing and characterization of specific T cell immunosurveillance in Kenyan children. Mol. Cell. Biol. 19, 1651–1660 (1999).
a clinical isolate of Epstein–Barr virus from J. Infect. Dis. 195, 799–808 (2007). 70. Komano, J., Maruo, S., Kurozumi, K., Oda, T. &
nasopharyngeal carcinoma tissue by using next- 47. Moormann, A. M., Snider, C. J. & Chelimo, K. The Takada, K. Oncogenic role of Epstein–Barr virus-
generation sequencing technology. J. Virol. 85, company malaria keeps: how co‑infection with Epstein– encoded RNAs in Burkitt’s lymphoma cell line Akata.
11291–11299 (2011). Barr virus leads to endemic Burkitt lymphoma. Curr. J. Virol. 73, 9827–9831 (1999).
23. Kwok, H. et al. Genomic sequencing and comparative Opin. Infect. Dis. 24, 435–441 (2011). 71. Jochner, N. et al. Epstein–Barr virus nuclear antigen 2
analysis of Epstein–Barr virus genome isolated from 48. Horne-Debets, J. M. et al. PD‑1 dependent exhaustion is a transcriptional suppressor of the immunoglobulin
primary nasopharyngeal carcinoma biopsy. PLoS ONE of CD8+ T cells drives chronic malaria. Cell Rep. 5, mu gene: implications for the expression of the
7, e36939 (2012). 1204–1213 (2013). translocated c‑myc gene in Burkitt’s lymphoma cells.
24. Lin, Z. et al. Whole-genome sequencing of the Akata 49. Moss, D. J. et al. A comparison of Epstein–Barr virus- EMBO J. 15, 375–382 (1996).
and Mutu Epstein–Barr virus strains. J. Virol. 87, specific T‑cell immunity in malaria-endemic and 72. Pajic, A. et al. Antagonistic effects of c‑myc and
1172–1182 (2013). -nonendemic regions of Papua New Guinea. Epstein–Barr virus latent genes on the phenotype
25. Tsai, M. H. et al. Spontaneous lytic replication and Int. J. Cancer 31, 727–732 (1983). of human B cells. Int. J. Cancer 93, 810–816
epitheliotropism define an Epstein–Barr virus strain 50. Njie, R. et al. The effects of acute malaria on Epstein– (2001).
found in carcinomas. Cell Rep. 5, 458–470 (2013). Barr virus (EBV) load and EBV-specific T cell immunity 73. Kelly, G., Bell, A. & Rickinson, A. Epstein–Barr virus-
26. Lei, H. et al. Identification and characterization of EBV in Gambian children. J. Infect. Dis. 199, 31–38 associated Burkitt lymphomagenesis selects for
genomes in spontaneously immortalized human (2009). downregulation of the nuclear antigen EBNA2. Nat.
peripheral blood B lymphocytes by NGS technology. 51. Torgbor, C. et al. A multifactorial role for P. falciparum Med. 8, 1098–1104 (2002).
BMC Genomics 14, 804 (2013). malaria in endemic Burkitt’s lymphoma pathogenesis. 74. Kelly, G. L. et al. Epstein–Barr virus nuclear
27. Kwok, H. et al. Genomic diversity of Epstein–Barr PLoS Pathog. 10, e1004170 (2014). antigen 2 (EBNA2) gene deletion is consistently
virus genomes isolated from primary nasopharyngeal 52. Wilmore, J. R. et al. AID expression in peripheral linked with EBNA3A, -3B, and -3C expression in
carcinoma biopsy samples. J. Virol. 88, blood of children living in a malaria holoendemic Burkitt’s lymphoma cells and with increased
10662–10672 (2014). region is associated with changes in B cell subsets and resistance to apoptosis. J. Virol. 79, 10709–10717
28. Santpere, G. et al. Genome-wide analysis of wild-type Epstein–Barr virus. Int. J. Cancer 136, 1371–1380 (2005).
Epstein–Barr virus genomes derived from healthy (2015). 75. Anderton, E. et al. Two Epstein–Barr virus (EBV)
individuals of the 1,000 Genomes project. Genome 53. MacNeil, A., Sumba, O. P., Lutzke, M. L., oncoproteins cooperate to repress expression of the
Biol. Evol. 6, 846–860 (2014). Moormann, A. & Rochford, R. Activation of the proapoptotic tumour-suppressor Bim: clues to the
29. Palser, A. L. et al. Genome diversity of Epstein–Barr Epstein–Barr virus lytic cycle by the latex of the plant pathogenesis of Burkitt’s lymphoma. Oncogene 27,
virus from multiple tumor types and normal infection. Euphorbia tirucalli. Br. J. Cancer 88, 1566–1569 421–433 (2008).
J. Virol. 89, 5222–5237 (2015). (2003). 76. Kelly, G. L. et al. An Epstein–Barr virus anti-apoptotic
30. Rowe, M. et al. Differences in B cell growth phenotype 54. Mannucci, S. et al. EBV reactivation and chromosomal protein constitutively expressed in transformed
reflect novel patterns of Epstein–Barr virus latent polysomies: Euphorbia tirucalli as a possible cofactor cells and implicated in Burkitt lymphomagenesis:
gene expression in Burkitt’s lymphoma cells. EMBO J. in endemic Burkitt lymphoma. Adv. Hematol. 2012, the Wp/BHRF1 link. PLoS Pathog. 5, e1000341
6, 2743–2751 (1987). 149780 (2012). (2009).

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

77. Tierney, R. J., Shannon-Lowe, C. D., Fitzsimmons, L., 98. Rickinson, A. B., Moss, D. J. & Pope, J. H. Long-term 120. Oyama, T. et al. Age-related EBV-associated B‑cell
Bell, A. I. & Rowe, M. Unexpected patterns of C‑cell-mediated immunity to Epstein–Barr virus in lymphoproliferative disorders constitute a distinct
Epstein–Barr virus transcription revealed by a high man. II. Components necessary for regression in clinicopathologic group: a study of 96 patients. Clin.
throughput PCR array for absolute quantification of virus-infected leukocyte cultures. Int. J. Cancer 23, Cancer Res. 13, 5124–5132 (2007).
viral mRNA. Virology 474, 117–130 (2015). 610–617 (1979). 121. Purtilo, D. T., Cassel, C. K., Yang, J. P. & Harper, R.
78. Henle, G., Henle, W. & Diehl, V. Relation of Burkitt’s 99. Moss, D. J. et al. Cytotoxic T‑cell clones discriminate X‑Linked recessive progressive combined variable
tumor-associated herpes-type virus to infectious between A- and B‑type Epstein–Barr virus immunodeficiency (Duncan’s disease). Lancet 1,
mononucleosis. Proc. Natl Acad. Sci. USA 59, 94–101 transformants. Nature 331, 719–721 (1988). 935–940 (1975).
(1968). 100. Burrows, S. R., Sculley, T. B., Misko, I. S., Schmidt, C. 122. Seemayer, T. A. et al. X‑Linked lymphoproliferative
79. Niederman, J. C., Miller, G., Pearson, H. A., & Moss, D. J. An Epstein–Barr virus-specific cytotoxic disease: twenty-five years after the discovery. Pediatr.
Pagano, J. S. & Dowaliby, J. M. Infectious T cell epitope in EBV nuclear antigen 3 (EBNA 3). Res. 38, 471–478 (1995).
mononucleosis. Epstein–Barr-virus shedding in J. Exp. Med. 171, 345–349 (1990). 123. Coffey, A. J. et al. Host response to EBV infection in
saliva and the oropharynx. N. Engl. J. Med. 294, 101. Kanekiyo, M. et al. Rational design of an Epstein–Barr X‑linked lymphoproliferative disease results from
1355–1359 (1976). virus vaccine targeting the receptor-binding site. Cell mutations in an SH2‑domain encoding gene. Nat.
80. Greenspan, J. S. et al. Replication of Epstein–Barr 162, 1090–1100 (2015). Genet. 20, 129–135 (1998).
virus within the epithelial cells of oral “hairy” 102. Hui, E. P. et al. Phase I trial of recombinant modified 124. Nichols, K. E. et al. Inactivating mutations in an SH2
leukoplakia, an AIDS-associated lesion. N. Engl. vaccinia ankara encoding Epstein–Barr viral tumor domain-encoding gene in X‑linked lymphoproliferative
J. Med. 313, 1564–1571 (1985). antigens in nasopharyngeal carcinoma patients. syndrome. Proc. Natl Acad. Sci. USA 95,
81. Babcock, G. J., Decker, L. L., Volk, M. & Thorley- Cancer Res. 73, 1676–1688 (2013). 13765–13770 (1998).
Lawson, D. A. EBV persistence in memory B cells 103. Sokal, E. M. et al. Recombinant gp350 vaccine for 125. Sayos, J. et al. The X‑linked lymphoproliferative-disease
in vivo. Immunity 9, 395–404 (1998). infectious mononucleosis: a phase 2, randomized, gene product SAP regulates signals induced through
82. Kurth, J. et al. EBV-infected B cells in infectious double-blind, placebo-controlled trial to evaluate the the co‑receptor SLAM. Nature 395, 462–469 (1998).
mononucleosis: viral strategies for spreading in the safety, immunogenicity, and efficacy of an Epstein– 126. Hislop, A. D. et al. Impaired Epstein–Barr virus-specific
B cell compartment and establishing latency. Immunity Barr virus vaccine in healthy young adults. J. Infect. CD8+ T‑cell function in X‑linked lymphoproliferative
13, 485–495 (2000). Dis. 196, 1749–1753 (2007). disease is restricted to SLAM family-positive B‑cell
83. Kurth, J., Hansmann, M. L., Rajewsky, K. & 104. Doak, P. B., Montgomerie, J. Z., North, J. D. & targets. Blood 116, 3249–3257 (2010).
Kuppers, R. Epstein–Barr virus-infected B cells Smith, F. Reticulum cell sarcoma after renal 127. MacMahon, B. Epidemiology of Hodgkin’s disease.
expanding in germinal centers of infectious homotransplantation and azathioprine and prednisone Cancer Res. 26, 1189–1201 (1966).
mononucleosis patients do not participate in the therapy. Br. Med. J. 4, 746–748 (1968). 128. Correa, P. & O’Conor, G. T. Epidemiologic patterns of
germinal center reaction. Proc. Natl Acad. Sci. USA 105. Penn, I., Hammond, W., Brettschneider, L. & Hodgkin’s disease. Int. J. Cancer 8, 192–201 (1971).
100, 4730–4735 (2003). Starzl, T. E. Malignant lymphomas in transplantation 129. Connelly, R. R. & Christine, B. W. A cohort study of
84. Thorley-Lawson, D. A. & Gross, A. Persistence of the patients. Transplant. Proc. 1, 106–112 (1969). cancer following infectious mononucleosis. Cancer Res.
Epstein–Barr virus and the origins of associated 106. Nagington, J. & Gray, J. Cyclosporin A 34, 1172–1178 (1974).
lymphomas. N. Engl. J. Med. 350, 1328–1337 immunosuppression, Epstein–Barr antibody, and 130. Rosdahl, N., Larsen, S. O. & Clemmesen, J. Hodgkin’s
(2004). lymphoma. Lancet 1, 536–537 (1980). disease in patients with previous infectious
85. Gires, O. et al. Latent membrane protein 1 of 107. Crawford, D. H. et al. Epstein Barr virus nuclear mononucleosis: 30 years’ experience. Br. Med. J. 2,
Epstein–Barr virus mimics a constitutively active antigen positive lymphoma after cyclosporin A 253–256 (1974).
receptor molecule. EMBO J. 16, 6131–6140 treatment in patient with renal allograft. Lancet 1, 131. Poppema, S., van Imhoff, G., Torensma, R. & Smit, J.
(1997). 1355–1356 (1980). Lymphadenopathy morphologically consistent with
86. Caldwell, R. G., Wilson, J. B., Anderson, S. J. & 108. Hanto, D. W., Sakamoto, K., Purtilo, D. T., Hodgkin’s disease associated with Epstein–Barr virus
Longnecker, R. Epstein–Barr virus LMP2A drives Simmons, R. L. & Najarian, J. S. The Epstein–Barr infection. Am. J. Clin. Pathol. 84, 385–390 (1985).
B cell development and survival in the absence of virus in the pathogenesis of posttransplant 132. Weiss, L. M., Strickler, J. G., Warnke, R. A.,
normal B cell receptor signals. Immunity 9, 405–411 lymphoproliferative disorders. Clinical, pathologic, and Purtilo, D. T. & Sklar, J. Epstein–Barr viral DNA in
(1998). virologic correlation. Surgery 90, 204–213 (1981). tissues of Hodgkin’s disease. Am. J. Pathol. 129,
87. Babcock, G. J., Hochberg, D. & Thorley-Lawson, A. D. 109. Saemundsen, A. K. et al. Documentation of Epstein– 86–91 (1987).
The expression pattern of Epstein–Barr virus latent Barr virus infection in immunodeficient patients with 133. Weiss, L. M., Movahed, L. A., Warnke, R. A. & Sklar, J.
genes in vivo is dependent upon the differentiation life-threatening lymphoproliferative diseases by Detection of Epstein–Barr viral genomes in Reed–
stage of the infected B cell. Immunity 13, 497–506 Epstein–Barr virus complementary RNA/DNA and Sternberg cells of Hodgkin’s disease. N. Engl. J. Med.
(2000). viral DNA/DNA hybridization. Cancer Res. 41, 320, 502–506 (1989).
88. Thorley-Lawson, D. A. Epstein–Barr virus: exploiting 4237–4242 (1981). 134. Wu, T. C. et al. Detection of EBV gene expression in
the immune system. Nat. Rev. Immunol. 1, 75–82 110. Hanto, D. W. et al. Epstein–Barr virus (EBV) induced Reed–Sternberg cells of Hodgkin’s disease. Int.
(2001). polyclonal and monoclonal B‑cell lymphoproliferative J. Cancer 46, 801–804 (1990).
89. Panagopoulos, D., Victoratos, P., Alexiou, M., diseases occurring after renal transplantation. Clinical, 135. Kuppers, R. et al. Hodgkin disease: Hodgkin and
Kollias, G. & Mosialos, G. Comparative analysis of pathologic, and virologic findings and implications for Reed–Sternberg cells picked from histological sections
signal transduction by CD40 and the Epstein–Barr therapy. Ann. Surg. 198, 356–369 (1983). show clonal immunoglobulin gene rearrangements
virus oncoprotein LMP1 in vivo. J. Virol. 78, 111. Schubach, W. H., Hackman, R., Neiman, P. E., and appear to be derived from B cells at various
13253–13261 (2004). Miller, G. & Thomas, E. D. A monoclonal stages of development. Proc. Natl Acad. Sci. USA 91,
90. Swanson-Mungerson, M. A., Caldwell, R. G., immunoblastic sarcoma in donor cells bearing 10962–10966 (1994).
Bultema, R. & Longnecker, R. Epstein–Barr virus Epstein–Barr virus genomes following allogeneic 136. Brauninger, A. et al. Molecular biology of Hodgkin’s
LMP2A alters in vivo and in vitro models of B‑cell marrow grafting for acute lymphoblastic leukemia. and Reed/Sternberg cells in Hodgkin’s lymphoma. Int.
anergy, but not deletion, in response to autoantigen. Blood 60, 180–187 (1982). J. Cancer 118, 1853–1861 (2006).
J. Virol. 79, 7355–7362 (2005). 112. Dotti, G. et al. Lymphomas occurring late after solid- 137. Chaganti, S. et al. Epstein–Barr virus infection in vitro
91. Thorley-Lawson, D. A., Duca, K. A. & Shapiro, M. organ transplantation: influence of treatment on the can rescue germinal center B cells with inactivated
Epstein–Barr virus: a paradigm for persistent infection clinical outcome. Transplantation 74, 1095–1102 immunoglobulin genes. Blood 106, 4249–4252
— for real and in virtual reality. Trends Immunol. 29, (2002). (2005).
195–201 (2008). 113. Rea, D. et al. Epstein–Barr virus latent and replicative 138. Mancao, C., Altmann, M., Jungnickel, B. &
92. Roughan, J. E. & Thorley-Lawson, D. A. The gene expression in post-transplant lymphoproliferative Hammerschmidt, W. Rescue of “crippled” germinal
intersection of Epstein–Barr virus with the germinal disorders and AIDS-related non-Hodgkin’s lymphomas. center B cells from apoptosis by Epstein–Barr virus.
center. J. Virol. 83, 3968–3976 (2009). French Study Group of Pathology for HIV-associated Blood 106, 4339–4344 (2005).
93. Roughan, J. E., Torgbor, C. & Thorley-Lawson, D. A. Tumors. Ann. Oncol. 5 (Suppl. 1), 113–116 (1994). 139. Bechtel, D., Kurth, J., Unkel, C. & Kuppers, R.
Germinal center B cells latently infected with Epstein– 114. Kanakry, J. A. & Ambinder, R. F. EBV-related Transformation of BCR-deficient germinal-center
Barr virus proliferate extensively but do not increase lymphomas: new approaches to treatment. Curr. Treat. B cells by EBV supports a major role of the virus in the
in number. J. Virol. 84, 1158–1168 (2010). Opt. Oncol. 14, 224–236 (2013). pathogenesis of Hodgkin and posttransplantation
94. Laichalk, L. L. & Thorley-Lawson, D. A. Terminal 115. Simmons, R. L. & Najarian, J. S. Immunosuppression lymphomas. Blood 106, 4345–4350 (2005).
differentiation into plasma cells initiates the and malignant neoplasms. N. Engl. J. Med. 283, 140. Pallesen, G., Hamilton-Dutoit, S. J., Rowe, M. &
replicative cycle of Epstein–Barr virus in vivo. J. Virol. 934–935 (1970). Young, L. S. Expression of Epstein–Barr virus latent
79, 1296–1307 (2005). 116. Starzl, T. E. et al. Reversibility of lymphomas and gene products in tumour cells of Hodgkin’s disease.
95. Sheldon, P. J., Hemsted, E. H., Papamichail, M. & lymphoproliferative lesions developing under Lancet 337, 320–322 (1991).
Holborow, E. J. Thymic origin of atypical lymphoid cells cyclosporin-steroid therapy. Lancet 1, 583–587 (1984). 141. Murray, P. G., Young, L. S., Rowe, M. & Crocker, J.
in infectious mononucleosis. Lancet 1, 1153–1155 117. Rooney, C. M. et al. Use of gene-modified virus-specific Immunohistochemical demonstration of the Epstein–
(1973). T lymphocytes to control Epstein–Barr-virus-related Barr virus-encoded latent membrane protein in
96. Pattengale, P. K., Smith, R. W. & Perlin, E. Atypical lymphoproliferation. Lancet 345, 9–13 (1995). paraffin sections of Hodgkin’s disease. J. Pathol. 166,
lymphocytes in acute infectious mononucleosis. 118. Heslop, H. E. et al. Long-term restoration of immunity 1–5 (1992).
Identification by multiple T and B lymphocyte against Epstein–Barr virus infection by adoptive 142. Deacon, E. M. et al. Epstein–Barr virus and Hodgkin’s
markers. N. Engl. J. Med. 291, 1145–1148 transfer of gene-modified virus-specific T lymphocytes. disease: transcriptional analysis of virus latency in the
(1974). Nat. Med. 2, 551–555 (1996). malignant cells. J. Exp. Med. 177, 339–349 (1993).
97. Moss, D. J., Rickinson, A. B. & Pope, J. H. Long-term 119. Bollard, C. M. et al. Sustained complete responses in 143. Murray, P. G., Constandinou, C. M., Crocker, J.,
T‑cell-mediated immunity to Epstein–Barr virus in patients with lymphoma receiving autologous Young, L. S. & Ambinder, R. F. Analysis of major
man. I. Complete regression of virus-induced cytotoxic T lymphocytes targeting Epstein–Barr virus histocompatibility complex class I, TAP expression, and
transformation in cultures of seropositive donor latent membrane proteins. J. Clin. Oncol. 32, LMP2 epitope sequence in Epstein–Barr virus-positive
leukocytes. Int. J. Cancer 22, 662–668 (1978). 798–808 (2014). Hodgkin’s disease. Blood 92, 2477–2483 (1998).

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

144. Alber, G. et al. Molecular mimicry of the antigen 169. Wolf, H., zur Hausen, H. & Becker, V. EB viral genomes 193. Miettinen, M. Smooth muscle tumors of soft tissue
receptor signalling motif by transmembrane proteins in epithelial nasopharyngeal carcinoma cells. Nat. New and non-uterine viscera: biology and prognosis. Mod.
of the Epstein–Barr virus and the bovine leukaemia Biol. 244, 245–247 (1973). Pathol. 27 (Suppl. 1), 17–29 (2014).
virus. Curr. Biol. 3, 333–339 (1993). 170. Pathmanathan, R., Prasad, U., Sadler, R., Flynn, K. & 194. Fernandez-Menendez, S., Fernandez-Moran, M.,
145. Mosialos, G. et al. The Epstein–Barr virus Raab-Traub, N. Clonal proliferations of cells infected Fernandez-Vega, I., Perez-Alvarez, A. & Villafani-
transforming protein LMP1 engages signaling proteins with Epstein–Barr virus in preinvasive lesions related Echazu, J. Epstein–Barr virus and multiple sclerosis.
for the tumor necrosis factor receptor family. Cell 80, to nasopharyngeal carcinoma. N. Engl. J. Med. 333, From evidence to therapeutic strategies. J. Neurol.
389–399 (1995). 693–698 (1995). Sci. 361, 213–219 (2016).
146. Hjalgrim, H. et al. Infectious mononucleosis, childhood 171. Fahraeus, R. et al. Expression of Epstein–Barr virus- 195. Imai, S., Nishikawa, J. & Takada, K. Cell‑to‑cell
social environment, and risk of Hodgkin lymphoma. encoded proteins in nasopharyngeal carcinoma. Int. contact as an efficient mode of Epstein–Barr virus
Cancer Res. 67, 2382–2388 (2007). J. Cancer 42, 329–338 (1988). infection of diverse human epithelial cells. J. Virol. 72,
147. Hjalgrim, H. et al. HLA‑A alleles and infectious 172. Brooks, L., Yao, Q. Y., Rickinson, A. B. & Young, L. S. 4371–4378 (1998).
mononucleosis suggest a critical role for cytotoxic Epstein–Barr virus latent gene transcription in 196. Shannon-Lowe, C. & Rowe, M. Epstein–Barr virus
T‑cell response in EBV-related Hodgkin lymphoma. nasopharyngeal carcinoma cells: coexpression of infection of polarized epithelial cells via the basolateral
Proc. Natl Acad. Sci. USA 107, 6400–6405 (2010). EBNA1, LMP1, and LMP2 transcripts. J. Virol. 66, surface by memory B cell-mediated transfer infection.
148. Diepstra, A. et al. Association with HLA class I in 2689–2697 (1992). PLoS Pathog. 7, e1001338 (2011).
Epstein–Barr-virus-positive and with HLA class III in 173. Busson, P. et al. Consistent transcription of the 197. Ni, C. et al. In‑cell infection: a novel pathway for
Epstein–Barr-virus-negative Hodgkin’s lymphoma. Epstein–Barr virus LMP2 gene in nasopharyngeal Epstein–Barr virus infection mediated by cell‑in‑cell
Lancet 365, 2216–2224 (2005). carcinoma. J. Virol. 66, 3257–3262 (1992). structures. Cell Res. 25, 785–800 (2015).
149. Doll, D. C. & List, A. F. Burkitt’s lymphoma in a 174. Niedobitek, G. et al. Epstein–Barr virus and 198. Kanda, T., Furuse, Y., Oshitani, H. & Kiyono, T. Highly
homosexual. Lancet 1, 1026–1027 (1982). carcinomas: undifferentiated carcinomas but not efficient CRISPR/Cas9‑mediated cloning and functional
150. Ziegler, J. L. et al. Outbreak of Burkitt’s‑like lymphoma squamous cell carcinomas of the nasopharynx are characterization of gastric cancer-derived Epstein–
in homosexual men. Lancet 2, 631–633 (1982). regularly associated with the virus. J. Pathol. 165, Barr virus strains. J. Virol. 90, 4383–4393 (2016).
151. Chaganti, R. S. et al. Specific translocations 17–24 (1991). 199. Nakatsuka, S. et al. Pyothorax-associated lymphoma:
characterize Burkitt’s‑like lymphoma of homosexual 175. Sheen, T. S., Tsai, C. C., Ko, J. Y., Chang, Y. L. & a review of 106 cases. J. Clin. Oncol. 20, 4255–4260
men with the acquired immunodeficiency syndrome. Hsu, M. M. Undifferentiated carcinoma of the major (2002).
Blood 61, 1265–1268 (1983). salivary glands. Cancer 80, 357–363 (1997). 200. Dojcinov, S. D., Venkataraman, G., Raffeld, M.,
152. Moir, S. & Fauci, A. S. Insights into B cells and HIV- 176. Lo, K. W., Chung, G. T. & To, K. F. Deciphering the Pittaluga, S. & Jaffe, E. S. EBV positive mucocutaneous
specific B‑cell responses in HIV-infected individuals. molecular genetic basis of NPC through molecular, ulcer—a study of 26 cases associated with various
Immunol. Rev. 254, 207–224 (2013). cytogenetic, and epigenetic approaches. Semin. sources of immunosuppression. Am. J. Surg. Pathol.
153. Haas, A., Zimmermann, K. & Oxenius, A. Antigen- Cancer Biol. 22, 79–86 (2012). 34, 405–417 (2010).
dependent and -independent mechanisms of T and 177. Jia, W. H. & Qin, H. D. Non-viral environmental 201. Zhang, G. et al. Circulating Epstein–Barr virus
B cell hyperactivation during chronic HIV‑1 infection. risk factors for nasopharyngeal carcinoma: a microRNAs miR‑BART7 and miR‑BART13 as
J. Virol. 85, 12102–12113 (2011). systematic review. Semin. Cancer Biol. 22, 117–126 biomarkers for nasopharyngeal carcinoma diagnosis
154. Biggar, R. J. et al. Hodgkin lymphoma and (2012). and treatment. Int. J. Cancer 136, E301–E312 (2015).
immunodeficiency in persons with HIV/AIDS. Blood 178. Young, L. S. & Rickinson, A. B. Epstein–Barr virus: 202. Yang, X. et al. Epigenetic markers for noninvasive early
108, 3786–3791 (2006). 40 years on. Nat. Rev. Cancer 4, 757–768 (2004). detection of nasopharyngeal carcinoma by
155. Jones, J. F. et al. T‑Cell lymphomas containing 179. Lin, D. C. et al. The genomic landscape of methylation-sensitive high resolution melting. Int.
Epstein–Barr viral DNA in patients with chronic nasopharyngeal carcinoma. Nat. Genet. 46, 866–871 J. Cancer 136, E127–E135 (2015).
Epstein–Barr virus infections. N. Engl. J. Med. 318, (2014). 203. Wildeman, M. A. et al. Cytolytic virus activation
733–741 (1988). 180. Shibata, D. & Weiss, L. M. Epstein–Barr virus- therapy for Epstein–Barr virus-driven tumors. Clin.
156. Kikuta, H. et al. Epstein–Barr virus genome-positive associated gastric adenocarcinoma. Am. J. Pathol. Cancer Res. 18, 5061–5070 (2012).
T lymphocytes in a boy with chronic active EBV 140, 769–774 (1992). 204. Li, J. H. et al. Efficacy of targeted FasL in
infection associated with Kawasaki-like disease. 181. Global Cancer Observatory. Estimated cancer nasopharyngeal carcinoma. Mol. Ther. 8, 964–973
Nature 333, 455–457 (1988). incidence, mortality and prevalence worldwide in (2003).
157. Harabuchi, Y. et al. Epstein–Barr virus in nasal T‑cell 2012. Globocan 2012 Cancer Fact Sheet: Stomach 205. Lee, E. K. et al. Small molecule inhibition of Epstein–
lymphomas in patients with lethal midline granuloma. cancer. http://globocan.iarc.fr/old/FactSheets/cancers/ Barr virus nuclear antigen‑1 DNA binding activity
Lancet 335, 128–130 (1990). stomach-new.asp (2012). interferes with replication and persistence of the viral
158. Cohen, J. I. Optimal treatment for chronic active 182. Lee, J. H. et al. Clinicopathological and molecular genome. Antiviral Res. 104, 73–83 (2014).
Epstein–Barr virus disease. Pediatr. Transplant. 13, characteristics of Epstein–Barr virus-associated 206. Gianti, E., Messick, T. E., Lieberman, P. M. &
393–396 (2009). gastric carcinoma: a meta-analysis. J. Gastroenterol. Zauhar, R. J. Computational analysis of EBNA1
159. Au, W. Y. et al. Clinical differences between nasal and Hepatol. 24, 354–365 (2009). “druggability” suggests novel insights for Epstein–Barr
extranasal natural killer/T‑cell lymphoma: a study of 183. Murphy, G., Pfeiffer, R., Camargo, M. C. & virus inhibitor design. J. Comput. Aided Mol. Des. 30,
136 cases from the International Peripheral T‑Cell Rabkin, C. S. Meta-analysis shows that prevalence of 285–303 (2016).
Lymphoma Project. Blood 113, 3931–3937 (2009). Epstein–Barr virus-positive gastric cancer differs 207. Taylor, G. S. et al. A recombinant modified vaccinia
160. Chiang, A. K., Tao, Q., Srivastava, G. & Ho, F. C. Nasal based on sex and anatomic location. Gastroenterology ankara vaccine encoding Epstein–Barr virus (EBV)
NK- and T‑cell lymphomas share the same type of 137, 824–833 (2009). target antigens: a phase I trial in UK patients with
Epstein–Barr virus latency as nasopharyngeal 184. Lee, H. S., Chang, M. S., Yang, H. K., Lee, B. L. & EBV-positive cancer. Clin. Cancer Res. 20,
carcinoma and Hodgkin’s disease. Int. J. Cancer 68, Kim, W. H. Epstein–Barr virus-positive gastric 5009–5022 (2014).
285–290 (1996). carcinoma has a distinct protein expression profile in 208. Smith, C. et al. Effective treatment of metastatic forms
161. Coleman, C. B. et al. Epstein–Barr virus type 2 latently comparison with Epstein−Barr virus-negative of Epstein–Barr virus-associated nasopharyngeal
infects T cells, inducing an atypical activation carcinoma. Clin. Cancer Res. 10, 1698–1705 (2004). carcinoma with a novel adenovirus-based adoptive
characterized by expression of lymphotactic cytokines. 185. Schneider, B. G. et al. Loss of p16/CDKN2A tumor immunotherapy. Cancer Res. 72, 1116–1125 (2012).
J. Virol. 89, 2301–2312 (2015). suppressor protein in gastric adenocarcinoma is 209. Barton, E. S. et al. Herpesvirus latency confers
162. Ng, S. B. et al. Activated oncogenic pathways and associated with Epstein–Barr virus and anatomic symbiotic protection from bacterial infection. Nature
therapeutic targets in extranodal nasal-type NK/T cell location in the body of the stomach. Hum. Pathol. 31, 447, 326–329 (2007).
lymphoma revealed by gene expression profiling. 45–50 (2000). 210. Zimber, U. et al. Geographical prevalence of two types
J. Pathol. 223, 496–510 (2011). 186. Ushiku, T. et al. p73 gene promoter methylation in of Epstein–Barr virus. Virology 154, 56–66 (1986).
163. de Schryver, A. et al. Epstein–Barr virus-associated Epstein–Barr virus-associated gastric carcinoma. Int. 211. Rickinson, A. B., Young, L. S. & Rowe, M. Influence of
antibody patterns in carcinoma of the post-nasal J. Cancer 120, 60–66 (2007). the Epstein–Barr virus nuclear antigen EBNA 2 on the
space. Clin. Exp. Immunol. 5, 443–459 (1969). 187. van Rees, B. P. et al. Different pattern of allelic loss in growth phenotype of virus-transformed B cells.
164. Henle, W. et al. Antibodies to Epstein–Barr virus in Epstein–Barr virus-positive gastric cancer with J. Virol. 61, 1310–1317 (1987).
nasopharyngeal carcinoma, other head and neck emphasis on the p53 tumor suppressor pathway. Am. 212. Yates, J. L., Warren, N. & Sugden, B. Stable replication
neoplasms, and control groups. J. Natl Cancer Inst. J. Pathol. 161, 1207–1213 (2002). of plasmids derived from Epstein–Barr virus in various
44, 225–231 (1970). 188. Wang, K. et al. Whole-genome sequencing and mammalian cells. Nature 313, 812–815 (1985).
165. Henle, W., Ho, J. H., Henle, G., Chau, J. C. & comprehensive molecular profiling identify new driver 213. Wilson, J. B., Bell, J. L. & Levine, A. J. Expression of
Kwan, H. C. Nasopharyngeal carcinoma: significance mutations in gastric cancer. Nat. Genet. 46, 573–582 Epstein–Barr virus nuclear antigen‑1 induces B cell
of changes in Epstein–Barr virus-related antibody (2014). neoplasia in transgenic mice. EMBO J. 15,
patterns following therapy. Int. J. Cancer 20, 189. Cancer Genome Atlas Research Network. 3117–3126 (1996).
663–672 (1977). Comprehensive molecular characterization of gastric 214. Gruhne, B. et al. The Epstein–Barr virus nuclear
166. Zeng, Y. et al. Prospective studies on nasopharyngeal adenocarcinoma. Nature 513, 202–209 (2014). antigen‑1 promotes genomic instability via induction
carcinoma in Epstein–Barr virus IgA/VCA antibody- 190. zur Hausen, A. et al. Epstein–Barr virus in gastric of reactive oxygen species. Proc. Natl Acad. Sci. USA
positive persons in Wuzhou City, China. Int. J. Cancer carcinomas and gastric stump carcinomas: a late 106, 2313–2318 (2009).
36, 545–547 (1985). event in gastric carcinogenesis. J. Clin. Pathol. 57, 215. Rabson, M., Gradoville, L., Heston, L. & Miller, G. Non-
167. Mutirangura, A. et al. Epstein–Barr viral DNA in 487–491 (2004). immortalizing P3J‑HR‑1 Epstein–Barr virus: a deletion
serum of patients with nasopharyngeal carcinoma. 191. Corvalan, A. et al. Association of a distinctive strain of mutant of its transforming parent, Jijoye. J. Virol. 44,
Clin. Cancer Res. 4, 665–669 (1998). Epstein–Barr virus with gastric cancer. Int. J. Cancer 834–844 (1982).
168. Lo, Y. M. et al. Quantitative and temporal correlation 118, 1736–1742 (2006). 216. Sakai, T. et al. Functional replacement of the
between circulating cell-free Epstein–Barr virus DNA 192. Muhe, J. & Wang, F. Non-human primate intracellular region of the Notch1 receptor by Epstein–
and tumor recurrence in nasopharyngeal carcinoma. lymphocryptoviruses: past, present, and future. Curr. Barr virus nuclear antigen 2. J. Virol. 72, 6034–6039
Cancer Res. 59, 5452–5455 (1999). Top. Microbiol. Immunol. 391, 385–405 (2015). (1998).

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

217. Tomkinson, B., Robertson, E. & Kieff, E. Epstein–Barr carcinoma and their roles in gastric carcinogenesis. 256. Hislop, A. D. et al. A CD8+ T cell immune evasion
virus nuclear proteins EBNA‑3A and EBNA‑3C are J. Virol. 89, 5581–5591 (2015). protein specific to Epstein–Barr virus and its close
essential for B‑lymphocyte growth transformation. 237. Kang, D., Skalsky, R. L. & Cullen, B. R. EBV BART relatives in Old World primates. J. Exp. Med. 204,
J. Virol. 67, 2014–2025 (1993). microRNAs target multiple pro-apoptotic cellular 1863–1873 (2007).
218. Parker, G. A. et al. Epstein–Barr virus nuclear antigen genes to promote epithelial cell survival. PLoS Pathog. 257. Zuo, J. et al. The Epstein–Barr virus G‑protein-coupled
(EBNA)3C is an immortalizing oncoprotein with similar 11, e1004979 (2015). receptor contributes to immune evasion by targeting
properties to adenovirus E1A and papillomavirus E7. 238. Lei, T. et al. Targeting of DICE1 tumor suppressor by MHC class I molecules for degradation. PLoS Pathog.
Oncogene 13, 2541–2549 (1996). Epstein–Barr virus-encoded miR‑BART3* microRNA 5, e1000255 (2009).
219. White, R. E. et al. EBNA3B‑deficient EBV promotes in nasopharyngeal carcinoma. Int. J. Cancer 133, 258. Rowe, M. et al. Host shutoff during productive
B cell lymphomagenesis in humanized mice and is 79–87 (2013). Epstein–Barr virus infection is mediated by BGLF5
found in human tumors. J. Clin. Invest. 122, 239. Hsu, C. Y. et al. The Epstein–Barr virus-encoded and may contribute to immune evasion. Proc. Natl
1487–1502 (2012). microRNA MiR‑BART9 promotes tumor metastasis by Acad. Sci. USA 104, 3366–3371 (2007).
220. Allan, G. J., Inman, G. J., Parker, B. D., Rowe, D. T. targeting E‑cadherin in nasopharyngeal carcinoma. 259. Hong, G. K. et al. Epstein–Barr virus lytic infection
& Farrell, P. J. Cell growth effects of Epstein–Barr virus PLoS Pathog. 10, e1003974 (2014). contributes to lymphoproliferative disease in a SCID
leader protein. J. Gen. Virol. 73, 1547–1551 (1992). 240. Kanda, T. et al. Clustered microRNAs of the Epstein– mouse model. J. Virol. 79, 13993–14003 (2005).
221. Wang, D., Liebowitz, D. & Kieff, E. An EBV membrane Barr virus cooperatively downregulate an epithelial 260. Ma, S. D. et al. A new model of Epstein–Barr virus
protein expressed in immortalized lymphocytes cell-specific metastasis suppressor. J. Virol. 89, infection reveals an important role for early lytic viral
transforms established rodent cells. Cell 43, 831–840 2684–2697 (2015). protein expression in the development of lymphomas.
(1985). 241. Qiu, J., Smith, P., Leahy, L. & Thorley-Lawson, D. A. J. Virol. 85, 165–177 (2011).
222. Kaye, K. M., Izumi, K. M. & Kieff, E. Epstein–Barr virus The Epstein–Barr virus encoded BART mi­­RNAs 261. Sato, Y. et al. Degradation of phosphorylated p53 by
latent membrane protein 1 is essential for potentiate tumor growth in vivo. PLoS Pathog. 11, viral protein-ECS E3 ligase complex. PLoS Pathog. 5,
B‑lymphocyte growth transformation. Proc. Natl Acad. e1004561 (2015). e1000530 (2009).
Sci. USA 90, 9150–9154 (1993). 242. Cai, L. et al. Epstein–Barr virus-encoded microRNA 262. Chang, Y. H. et al. Epstein–Barr virus BGLF4 kinase
223. Dawson, C. W., Port, R. J. & Young, L. S. The role of BART1 induces tumour metastasis by regulating retards cellular S‑phase progression and induces
the EBV-encoded latent membrane proteins LMP1 PTEN-dependent pathways in nasopharyngeal chromosomal abnormality. PLoS ONE 7, e39217
and LMP2 in the pathogenesis of nasopharyngeal carcinoma. Nature Commun. 6, 7353 (2015). (2012).
carcinoma (NPC). Semin. Cancer Biol. 22, 144–153 243. Wong, A. M., Kong, K. L., Tsang, J. W., Kwong, D. L. & 263. Wu, C. C. et al. Epstein–Barr virus DNase (BGLF5)
(2012). Guan, X. Y. Profiling of Epstein–Barr virus-encoded induces genomic instability in human epithelial cells.
224. Longnecker, R. & Kieff, E. A second Epstein–Barr virus microRNAs in nasopharyngeal carcinoma reveals Nucleic Acids Res. 38, 1932–1949 (2010).
membrane protein (LMP2) is expressed in latent potential biomarkers and oncomirs. Cancer 118, 264. Lin, Z. et al. Quantitative and qualitative RNA–Seq-
infection and colocalizes with LMP1. J. Virol. 64, 698–710 (2012). based evaluation of Epstein–Barr virus transcription
2319–2326 (1990). 244. Feederle, R. et al. The members of an Epstein–Barr in type I latency Burkitt’s lymphoma cells. J. Virol. 84,
225. Pearson, G. R. et al. Identification of an Epstein–Barr virus microRNA cluster cooperate to transform B 13053–13058 (2010).
virus early gene encoding a second component of the lymphocytes. J. Virol. 85, 9801–9810 (2011). 265. Young, L. S. & Murray, P. G. Epstein–Barr virus and
restricted early antigen complex. Virology 160, 245. Rymo, L. Identification of transcribed regions of oncogenesis: from latent genes to tumours. Oncogene
151–161 (1987). Epstein–Barr virus DNA in Burkitt lymphoma-derived 22, 5108–5121 (2003).
226. Henderson, S. et al. Epstein–Barr virus-coded BHRF1 cells. J. Virol. 32, 8–18 (1979). 266. Lo, K. W. & Huang, D. P. Genetic and epigenetic
protein, a viral homologue of Bcl‑2, protects human 246. Glickman, J. N., Howe, J. G. & Steitz, J. A. Structural changes in nasopharyngeal carcinoma. Semin. Cancer
B cells from programmed cell death. Proc. Natl Acad. analyses of EBER1 and EBER2 ribonucleoprotein Biol. 12, 451–462 (2002).
Sci. USA 90, 8479–8483 (1993). particles present in Epstein–Barr virus-infected cells. 267. Tsang, C. M. et al. Cyclin D1 overexpression supports
227. Watanabe, A. et al. Epstein–Barr virus-encoded Bcl‑2 J. Virol. 62, 902–911 (1988). stable EBV infection in nasopharyngeal epithelial cells.
homologue functions as a survival factor in 247. Takada, K. Role of EBER and BARF1 in Proc. Natl Acad. Sci. USA 109, E3473–E3482 (2012).
Wp‑restricted Burkitt lymphoma cell line P3HR‑1. nasopharyngeal carcinoma (NPC) tumorigenesis. 268. Sheu, L. F. et al. Analysis of bcl‑2 expression in
J. Virol. 84, 2893–2901 (2010). Semin. Cancer Biol. 22, 162–165 (2012). normal, inflamed, dysplastic nasopharyngeal epithelia,
228. Wei, M. X. & Ooka, T. A transforming function of the 248. Fixman, E. D., Hayward, G. S. & Hayward, S. D. Trans- and nasopharyngeal carcinoma: association with p53
BARF1 gene encoded by Epstein–Barr virus. EMBO J. acting requirements for replication of Epstein–Barr expression. Hum. Pathol. 28, 556–562 (1997).
8, 2897–2903 (1989). virus ori-Lyt. J. Virol. 66, 5030–5039 (1992). 269. Chang, J. T. et al. Telomerase activity is frequently
229. Wei, M. X., Moulin, J. C., Decaussin, G., Berger, F. & 249. Schepers, A., Pich, D. & Hammerschmidt, W. found in metaplastic and malignant human
Ooka, T. Expression and tumorigenicity of the Epstein– Activation of oriLyt, the lytic origin of DNA replication nasopharyngeal tissues. Br. J. Cancer 82, 1946–1951
Barr virus BARF1 gene in human Louckes of Epstein–Barr virus, by BZLF1. Virology 220, (2000).
B‑lymphocyte cell line. Cancer Res. 54, 1843–1848 367–376 (1996). 270. Hao, D. et al. Evaluation of E‑cadherin, beta-catenin
(1994). 250. Zhang, Q. et al. Functional and physical interactions and vimentin protein expression using quantitative
230. Zhang, C. X., Decaussin, G., Daillie, J. & Ooka, T. between the Epstein–Barr virus (EBV) proteins BZLF1 immunohistochemistry in nasopharyngeal carcinoma
Altered expression of two Epstein–Barr virus early and BMRF1: effects on EBV transcription and lytic patients. Clin. Invest. Med. 37, E320–E330 (2014).
genes localized in BamHI‑A in nonproducer Raji cells. replication. J. Virol. 70, 5131–5142 (1996). 271. Li, L., Zhang, Y., Guo, B. B., Chan, F. K. & Tao, Q.
J. Virol. 62, 1862–1869 (1988). 251. Gao, Z. et al. The Epstein–Barr virus lytic Oncogenic induction of cellular high CpG methylation
231. Seto, E. et al. Epstein–Barr virus (EBV)-encoded transactivator Zta interacts with the helicase-primase by Epstein–Barr virus in malignant epithelial cells.
BARF1 gene is expressed in nasopharyngeal replication proteins. J. Virol. 72, 8559–8567 (1998). Chin. J. Cancer 33, 604–608 (2014).
carcinoma and EBV-associated gastric carcinoma 252. Wen, W. et al. Epstein–Barr virus BZLF1 gene, a
tissues in the absence of lytic gene expression. J. Med. switch from latency to lytic infection, is expressed as Acknowledgements
Virol. 76, 82–88 (2005). an immediate-early gene after primary infection of B The authors are grateful to A. Bell for help with revisions to
232. Pfeffer, S. et al. Identification of virus-encoded lymphocytes. J. Virol. 81, 1037–1042 (2007). the figures. L.S.Y. has been supported by Cancer Research
microRNAs. Science 304, 734–736 (2004). 253. Shannon-Lowe, C. et al. Features distinguishing UK. L.F.Y. is supported by High Impact Research Grant
233. Kozomara, A. & Griffiths-Jones, S. miRBase: Epstein–Barr virus infections of epithelial cells and UM.C/625/1/HIR/MOHE/DENT/23 from the University of
annotating high confidence microRNAs using deep B cells: viral genome expression, genome Malaya. P.G.M. is supported by a Bloodwise Programme
sequencing data. Nucleic Acids Res. 42, D68–D73 maintenance, and genome amplification. J. Virol. 83, Grant.
(2014). 7749–7760 (2009).
234. Marquitz, A. R. & Raab-Traub, N. The role of mi­­RNAs 254. Bhende, P. M., Seaman, W. T., Delecluse, H. J. & Competing interests statement
and EBV BARTs in NPC. Semin. Cancer Biol. 22, Kenney, S. C. The EBV lytic switch protein, Z, The authors declare no competing interests.
166–172 (2012). preferentially binds to and activates the methylated
235. Vereide, D. T. et al. Epstein–Barr virus maintains viral genome. Nat. Genet. 36, 1099–1104 (2004).
lymphomas via its mi­­RNAs. Oncogene 33, 255. Ma, S. D. et al. An Epstein–Barr virus (EBV) mutant DATABASES
1258–1264 (2014). with enhanced BZLF1 expression causes lymphomas GenBank: http://www.ncbi.nlm.nih.gov/nuccore/NC_007605
236. Shinozaki-Ushiku, A. et al. Profiling of virus-encoded with abortive lytic EBV infection in a humanized ALL LINKS ARE ACTIVE IN THE ONLINE PDF
microRNAs in Epstein–Barr virus-associated gastric mouse model. J. Virol. 86, 7976–7987 (2012).

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like