You are on page 1of 9

Oral Oncology 50 (2014) 330–338

Contents lists available at ScienceDirect

Oral Oncology
journal homepage: www.elsevier.com/locate/oraloncology

Etiological factors of nasopharyngeal carcinoma


Sai Wah Tsao a,⇑, Yim Ling Yip a, Chi Man Tsang a, Pei Shin Pang a, Victoria Ming Yi Lau a, Guitao Zhang a,
Kwok Wai Lo b,⇑
a
Department of Anatomy and Center for Cancer Research, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
b
Department of Anatomical and Cellular Pathology and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region

a r t i c l e i n f o s u m m a r y

Article history: Nasopharyngeal carcinoma (NPC) is a common disease among southern Chinese. The major etiological
Available online 12 March 2014 factors proposed for NPC pathogenesis include genetic susceptibility, environment factors and EBV infec-
tion. In the high risk population, genetic susceptibility to NPC has been mapped to the HLA loci and adja-
Keywords: cent genes in MHC region on chromosome 6p21. Consumption of preserved food including salted fish has
EBV been implicated in its etiology in earlier studies. Its contribution to pathogenesis of NPC remains to be
Nasopharyngeal carcinoma determined. A decreasing trend of NPC incidence was observed in Hong Kong, Taiwan and Singapore
Etiology
in recent years which may be accounted by a change of dietary habits. A comprehensive epidemiological
Stromal inflammation
Immune evasion
study will help to elucidate the relative importance of various risk factors in the pathogenesis of NPC.
Despite the close association of EBV infection with NPC, the etiological role of EBV in NPC pathogenesis
remains enigmatic. EBV infection in primary nasopharyngeal epithelial cells is uncommon and difficult to
achieve. EBV does not transform primary nasopharyngeal epithelial cells into proliferative clones, which
contrasts greatly with the well-documented ability of EBV to transform and immortalize primary B cells.
Genetic alterations identified in premalignant nasopharyngeal epithelium may play crucial roles to sup-
port stable EBV infection. Subsequently, latent and lytic EBV gene products may drive clonal expansion
and transformation of premalignant nasopharyngeal epithelial cells into cancer cells. Stromal inflamma-
tion in nasopharyngeal mucosa is believed to play an important role in modulating the growth and pos-
sibly drive the malignant transformation of EBV-infected nasopharyngeal epithelial cells. Furthermore,
there are increasing evidences supporting a role of EBV infection to evade host immune surveillance.
EBV-infected cells may have selective growth advantages in vivo by acquiring a stress–resistance pheno-
type. Understanding the etiological factors and pathogenesis of NPC will contribute effectively to the pre-
vention and treatment of this disease.
Ó 2014 Elsevier Ltd. All rights reserved.

Geographical distribution of nasopharyngeal carcinoma also seen in regions in North Africa. Globally, approximately
65,000 new cases of NPC are reported each year and more than
Nasopharyngeal carcinoma (NPC) stands out from other epithe- 80% are from southern China and Southeast Asia. The highest inci-
lial cancers arising from the head and neck regions in its distinct dence rate of NPC is amongst Cantonese living in Hong Kong and
geographical distribution and close association with EBV infection Guang-dong province of Southern China (>20 per 100,000 per year
[1,2]. The anatomic site of NPC is also unique which may implicate in males) [3–5]. Relatively high incidence of NPC was also reported
a contributing role of microenvironment in its pathogenesis. The in other provinces of Southern China region including GuangXi,
incidence rate of NPC is remarkably variable worldwide. In most Fujian and Hunan. The incidence rate of NPC is low in Northern
parts of the world, NPC incidence is low (<1/100,000 per year). China (1 to 5 per 100,000 persons per year). Notably, southern
NPC incidence is high among ethnic southern Chinese and native Chinese migrated to non-endemic regions still retains a much
Eskimos living in Greenland and Alaska. High incidence of NPC is higher incidence rate compared to other ethnic groups living in
the same geographical regions. Among the high-risk population,
incidence of NPC peaks between 45 and 54 years old and declines
⇑ Corresponding authors. Address: Laboratory Block, Department of Anatomy,
thereafter. For unknown reason, men are roughly 2–3 folds more
LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special
frequently affected than women [3–5]. The remarkable racial and
Administrative Region. Tel.: +852 28199227; fax: +852 28170857 (S.W. Tsao). geographic distribution of NPC suggests a strong association of
E-mail addresses: gswtsao@hku.hk (S.W. Tsao), kwlo@cuhk.edu.hk (K.W. Lo). NPC with genetic susceptibility and environmental factors.

http://dx.doi.org/10.1016/j.oraloncology.2014.02.006
1368-8375/Ó 2014 Elsevier Ltd. All rights reserved.
S.W. Tsao et al. / Oral Oncology 50 (2014) 330–338 331

Interestingly, the incidence rate of NPC has been decreasing con- earlier, the incidence of NPC is 20–50 folds higher in southern Chi-
tinuously in Hong Kong from 1980 (28.5/100,000 in males and nese compared to populations in Western countries. Notably, the
11.2/100,000 in females) to 2009 (14.0/100,000 in males and 4.6/ secondary and third generations of the southern Chinese who emi-
100,000 in females), a mark decrease of over 50% in the past grated to low incidence area in the USA still retains a higher risk of
30 years [6,7]. The decreasing trend in this high-risk region may re- NPC than the resident population despite of cultural assimilation
flect the change of certain risk factors related to dietary habits and [16]. Familial clustering of the disease was observed at a frequency
lifestyle. The continuous immigration of Chinese from non-ende- around 10% among Chinese patients [17]. Sharing common envi-
mic region to Hong Kong may also contribute partly to the decreas- ronmental risk factors may also account for familial aggregation
ing incidence rate of NPC [8]. Similar decreasing trend was also of NPC and may be difficult to distinguish from inherited genetic
reported in Taiwan [9] and Singapore [10]. The decreasing trend susceptibility. Nonetheless, genetic susceptibility loci to NPC in
of incidence of NPC in the past 20–25 years was however not ob- high-risk population have been reported notably the association
served in other regions of Guangdong province, including Sihui with the HLA class I genes in the MHC locus at chromosome
and Cangwu counties, where traditional dietary habits were still 6p21 [18]. The HLA class I genes encode proteins to identify and
maintained until early 1990’s [11]. present foreign antigens, including EBV-encoded peptides, to the
cytotoxic T cells to trigger the host immune response against vi-
rally infected cells. The variable degree of susceptibility to NPC
Histopathology of NPC
among different ethnic populations may reflect the differential
ability of HLA haplotypes in controlling EBV infection in infected
The nasopharynx is a tubular space situated at the base of the
populations. Individuals with specific HLA alleles may be less effi-
skull. It represents a transitional area between the nasal cavities
cient in mounting a cytotoxic immune response against EBV-in-
and the oropharynx, forming part of the Waldeyer ring of lymphoid
fected cells and may contribute to increased susceptibility to
tissues. The nasopharyngeal mucosa shows numerous folds and
NPC. An early study in affected sib pairs from southern China iden-
crypts. It consists of a special type of stratified squamous epithe-
tified a NPC susceptible locus closely linked to the HLA region [19].
lium referred as intermediate or transitional epithelium [12]. Var-
A large scale genome-wide association studies (GWAS) reported
iable amount of mixed stratified squamous epithelium and ciliated
recently also revealed that genes within the HLA region on chro-
epithelium are present at the lateral and posterior wall of the naso-
mosome 6p21 are strongly associated with NPC [20]. Independent
pharynx. Aside from sero-mucous glands, abundant infiltration of
and strong association signals in the HLA-A and HLA-B/C loci were
lymphocytes are found in the underlying stroma of the nasopha-
observed. Other studies have also confirmed the consistent associ-
ryngeal epithelium. NPC is commonly developed from the lateral
ation of NPC risk with HLA regions [21]. However, several non-HLA
wall of nasopharynx, especially at the fossa of Rosenmüller and
genes including GABBR1 and MICA in the HLA region also show
superior posterior wall [13]. The WHO classification in 1978 recog-
strong association on NPC susceptibility [22,23]. Apart from the
nized three histological subtypes of nasopharyngeal carcinoma:
genes in HLA region, significant association with MDS1-EVI1 at
squamous cell carcinoma (WHO type 1), nonkeratinizing carci-
3q26.2, CDKN2A/2B at 9p21.3, and TNFRSF19 at 13q12.12 was also
noma (WHO type 2), and undifferentiated carcinoma (WHO type
reported [20]. Identification of the causal variants in these loci and
3) which was modified in 1991. The WHO type 1 NPC was retained,
elucidation of their functions should enhance our understanding
while the WHO type 2 and type 3 are combined into a single cate-
on the contribution of genetic factors in NPC pathogenesis.
gory of ‘‘nonkeratinizing carcinoma’’, which was further subdi-
In addition to GWAS, three pedigree-based linkage studies per-
vided as ‘‘differentiated’’ or ‘‘undifferentiated’’ [14]. The
formed in high-risk NPC Chinese families have also identified NPC
histological features of keratinizing squamous cell carcinoma of
associated genes with high penetrance. Using microsatellite poly-
NPC are similar to squamous cell carcinoma arising from other
morphic markers, three disease susceptibility loci on chromosomes
sites of the head and neck regions with well-differentiated histo-
3p21.3, 4p15.1-q12 and 5p13 were identified from high-risk NPC
logical features including presence of intercellular bridges, keratin
Chinese pedigrees [24–26]. However, the susceptibility locus iden-
production and epithelial pearl formation. NPC in the endemic
tified in each of these familial studies is diverse. These non-concor-
areas of Southern China is mainly of the nonkeratinizing carcinoma
dant findings may be due to genetic heterogeneity and limited
which occupies up to 99% of all cases. The nonkeratinizing carci-
number of the Chinese familial NPC cases in these studies. Fine-
noma lacks keratinization features and is ubiquitously associated
mapping and functional characterization of these susceptibility
with EBV infection [4,15].
genes are warranted.
Case-control studies have demonstrated a link between genetic
Etiological factors of NPC polymorphism and NPC risk by influencing the individual suscep-
tibility to EBV infection and/or chemical carcinogen-induced cell
The epidemiological studies reveal distinctive ethnic and geo- transformation. By candidate gene-based approach, increased risks
graphic distribution of NPC strongly indicating that genetic suscep- of NPC were shown to be associated with genetic polymorphisms
tibility plays a major contributing role. The aforementioned involved in nitrosamine metabolism (CYP2E1, CYP2A6), detoxify-
decreasing trends of NPC incidence reported in Hong Kong, Taiwan ing carcinogenic electrophiles (GSTM1), DNA repair (XRCC1,
and Singapore population indicate that alteration of dietary habits hOGG1, NBS1), EBV entry into nasopharyngeal epithelium (PIGR),
and change in environment factors may also alter incidence rate of interleukins (IL1A, IL1B, IL2, IL8 and IL10) and toll-like receptors
NPC. The clonal origin of EBV infection and its ubiquitous presence (TLR3, TLR4, TLR10) [27–36]. Significant correlation of variants of
in NPC strongly indicates its involvement in NPC pathogenesis [2]. cancer-associated genes, including TP53, MDM2, CCND1 and
EBV infection, together with genetic susceptibility and environ- NEDD4, was also reported to be associated with increased NPC risk
mental factors are considered as the three major etiological factors among southern Chinese [37–41].
of NPC among southern Chinese in endemic areas of NPC.
Environmental factors
Genetic susceptibility
A number of agents in the environment have been postulated to
The distinct ethnicity of NPC indicates the important contribu- be associated with NPC risk. It has been reported in earlier studies
tion of genetic susceptibility to the pathogenesis of NPC. As stated that the ingestion of Cantonese-style salted fish, especially during
332 S.W. Tsao et al. / Oral Oncology 50 (2014) 330–338

childhood, correlates with increased risk of NPC in endemic popu- viral gene expression profile distinctive of type II latency [65]. Anal-
lations [5,42–47]. Experimental studies have shown that nasal and ysis of the TR repeats in EBV episomes isolated from premalignant
paranasal malignant tumors were developed in rats fed with nasopharyngeal epithelium and NPC showed clonal origin of EBV
salted-fish [48,49]. Other salted and preserved food items includ- infection [66] suggesting that NPC is derived from the expansion
ing shrimp paste and preserved vegetables may represent indepen- of a single EBV-infected cell [67]. Using microdissected tissues, alle-
dent risk factors for NPC among Chinese [5,42–44]. Volatile lic deletion of 3p and 9p could be detected in premalignant and his-
nitrosamines in some of the traditional food items of southern Chi- tologically normal nasopharyngeal epithelial tissues free of EBV
nese were postulated as the putative carcinogens for NPC develop- infection suggesting that genetic alterations in premalignant naso-
ment [50,51]. The active carcinogenic metabolite of these pharyngeal epithelium are present prior to EBV infection [68,69].
nitrosamines may induce DNA damage and chronic inflammation Our recent study shows that genetic alterations (e.g. cyclin D1 over-
in nasopharyngeal mucosa of genetically susceptible individuals. expression) in premalignant or dysplastic nasopharyngeal epithe-
Consuming this dietary carcinogen during childhood may lead to lium supports persistence of EBV episomes in infected cells [70].
accumulation of aberrant genetic lesions and development of field
cancerization in nasopharynx at early age, predisposing them to EBV readily infects and transforms primary B cells but not primary
EBV infection and thereby increase the risk of developing NPC. nasopharyngeal epithelial cells
The consumption of salt fish among Hong Kong population has EBV infection is rarely detected in vivo in nasopharyngeal epi-
dropped dramatically for the past 20 years. It remains to be deter- thelium except in premalignant nasopharyngeal dysplastic lesions
mined if the change of dietary habits is accountable for the contin- and nonkeratinizing NPC [4]. Most children in the developing world
uous decreasing trend of NPC incidence reported in Hong Kong for are infected with EBV before the age of three and mostly asymp-
the past 30 years [6,7]. tomatic in nature [60]. EBV infects B lymphocyte through binding
The use of traditional herbal medicines has also been reported to the CR2 (CD21) receptor on the surfaces of B lymphocytes [71].
to be a unique factor associated with elevated NPC risk in Asian The CR2 receptor is generally low or absent in mucosal epithelium
populations [5,52–54]. Some Chinese herbal plants may contribute lining the head and neck regions as detected by immunocytochem-
to NPC development by inducing EBV lytic antigen expression istry. Nonetheless, mRNA for CR2 receptor could be detected in ton-
[55,56]. In contrast to the preserved foods and herbal medicines, sillar epithelial cells [72]. A low% (1 in 105 cells) of EBV-infected
consumption of fresh fruit and leafy vegetables, especially during epithelial cells was reported in ex vivo cultures of asymptomatic
childhood, has been reported to be protective factors against NPC EBV carriers [73]. EBV replication was reported in 1.3% of tongues
in Chinese [57]. mucosa [74]. These findings support the involvement of oropharyn-
Non-dietary risk factors associated with increased NPC risk geal epithelial cells in the production of infectious EBV in vivo.
have also been reported. Occupational exposures to formaldehyde, Events that facilitate entry and establishment of EBV infection in
wood dust, fumes and chemicals have been recognized as NPC risk human nasopharyngeal epithelial cells are only beginning to be
factors by causing chronic inflammation in nasopharynx understood [75]. Recent studies showed that human epithelial cells
[3,5,53,58]. Cigarette smoking has been reported to be correlated could be infected in vitro, through the formation of specialized B
with well-differentiated NPC in low-risk population; its association cell-epithelial cell conjugates involving the EBV viral envelope gly-
with undifferentiated or nonkeratinizing carcinoma in the endemic coprotein, GP350, and the CR2 receptor of B lymphocytes [76,77].
area is controversial [57]. This cell-to-cell method of EBV infection of nasopharyngeal epithe-
lium may take place in vivo. EBV-infected B cells invariably may be
Epstein–Barr virus (EBV) infection present in the nasopharyngeal mucosa and have chance to come
into close contact with the polarized nasopharyngeal epithelial
Despite the close association of EBV infection with NPC, the role cells to transmit the EBV. Cell-free EBV virions may also enter the
of EBV infection in NPC pathogenesis remains enigmatic. An intri- epithelial cells through the basolateral membranes involving inter-
cate interplay of EBV with host stroma and genetic alteration in in- action of the EBV glycoprotein, BMRF-2, with the integrin a5b1 of
fected host cells is likely to be involved in the pathogenesis of NPC the epithelial cells which may represent a transmission route of
(Fig. 1). EBV virus from infected epithelial cells across the lateral mem-
The association of EBV and human malignancies dated back to branes to infect adjacent epithelial cells [78]. EBV entry into epithe-
1960’s with the discovery of EBV infection in Burkitt’s lymphoma, lial cells may also be achieved via the IgA (against EBV-VCA)/Sc
a childhood cancer seen in Africa [59]. In vitro, EBV infection readily (secretory component) complex present on the surface of mucosal
drives proliferation and immortalization of B lymphocytes. EBV epithelial cells through endocytosis [79,80].
infection accounts for 90% of infectious mononucleosis, driving
the proliferation of infected B-lymphocytes in vivo [60,61]. The lym- Genetic alterations in premalignant nasopharyngeal epithelium
phoblastoid proliferation is self-limiting and will eventually subside support stable EBV infection
as the host-immune defense mechanisms are activated, driving EBV Different mechanisms are involved in facilitating entry of EBV
into latent infection in memory B-lymphocytes. EBV infection in into epithelial cells and persistence of EBV episomes post infection.
premalignant nasopharyngeal epithelial cells harboring genetic EBV infection of primary nasopharyngeal epithelial cells does not in-
alterations may drive their malignant transformation as proposed duce their proliferation, which contrast greatly to EBV infection in B-
in Burkitt’s lymphoma, Hodgkin’s disease and gastric carcinoma. lymphocytes [81]. However, infection of established epithelial cells
The association of EBV infection and NPC was first indicated harboring multiple genetic alterations may promote growth and
based on the serological evidence that NPC patients had higher confer tumorigenicity in infected cells [82]. In nasopharyngeal epi-
antibody titers against viral capsid antigen and early antigen than thelial cells immortalized by telomerase, EBV infection induces
healthy controls [62]. Elevated IgA and anti-DNase antibodies senescence and growth arrest phenotypes [70]. Overexpression of
against EBV were shown to have strong association with subse- cyclin D1 in the immortalized nasopharyngeal epithelial cells could
quent development of cancer in a large cohort [63]. A more recent overcome the growth arrest phenotype induced by EBV infection
study also showed that pretreatment levels of EBV DNase-specific and support long-term propagation of EBV-infection in infected
neutralizing antibody could be used as a prognostic marker to com- cells. Cyclin D1 overexpression is common in dysplastic nasopha-
plement TNM classification in nasopharyngeal carcinoma [64]. In ryngeal epithelium. Suppression of p16 expression also supports
almost all nonkeratinizing NPC, EBV genome could be detected with propagation of EBV infection in immortalized nasopharyngeal epi-
S.W. Tsao et al. / Oral Oncology 50 (2014) 330–338 333

Figure 1. Tumorigenesis model of pathogenesis of EBV-associated nasopharyngeal carcinoma.

thelial cells. Hence, genetic alterations in premalignant nasopharyn- cells in undifferentiated NPC from patients. Furthermore, NPC
geal epithelium which support EBV persistence of EBV may facilitate xenografts (xeno666, xeno2117, C15 and C17) which have been
their transformation into cancer cells. Counterintuitively, we failed passaged in immune deficient animals for over 20 years still retain
to observe growth advantage in EBV-infected nasopharyngeal epi- their EBV episomes. The nature of these selection advantages of
thelial cells or NPC cells in vitro [75]. It is also well-documented that EBV-infected NPC in vivo is unclear but may include (a) enhanced
EBV-infected NPC cells readily lost their EBV genomes upon propa- survival against apoptotic stimuli, (b) enhanced growth in inflam-
gation in culture showing that EBV infection per se does not confer matory stroma and (c) evasion from host immune surveillance.
growth advantage in vitro. The C666-1, which was established from Elucidation of growth advantages of EBV infection in NPC cells
a xenografted NPC passaged in nude mice for prolonged period, is in vivo may reveal the role of EBV in NPC pathogenesis.
the only NPC cell line which continues to harbor EBV upon prolonged
propagation. A defect in the lytic reactivation of EBV genome in
C666-1 cells and/or unique host factors supporting EBV persistence EBV infection confers a stress–resistant phenotype in infected cells.
may be involved. We were able to establish stable EBV infection in multiple lines of
immortalized nasopharyngeal epithelial cells and NPC cells [70].
The most consistent cellular phenotype observed is their resistance
Growth advantages of EBV-infected NPC cells in vivo to nutrient and growth factor deprivation. Compared to uninfected
In contrast to the rapid loss of EBV-infected NPC cells in vitro, cells, EBV-infected cells often reveal a robust phenotype in surviv-
EBV-infected NPC cells are actively selected and maintained ing nutrient deprivation. Detail mechanisms involved are not com-
in vivo. EBV episome could be detected in practically all cancer pletely elucidated but activation of PI3 K and Akt pathways are
334 S.W. Tsao et al. / Oral Oncology 50 (2014) 330–338

involved [75,83]. Stress–resistance phenotype could also be ob- tumorigenesis. EBV infection may support infected premalignant
served in nasopharyngeal epithelial cells expressing the EBV-en- or malignant nasopharyngeal epithelial cells to escape immuno-
coded LMP1 [84]. LMP1 expression was shown to inhibit the surveillance in vivo. Multiple mechanisms may be involved. Pre-
AMPK/LKB1 signaling pathways to promote cellular growth and sentation of antigenic peptides on MHC class I molecules is a
survival [85]. In EBV-infected cells, LMP1 expression could be in- signal for CD8 + T cells to recognize non-self antigens in defending
duced upon nutrient and growth factor deprivation to enhance cell parasite infection. EBV infection may suppress expression of MHC I
survival (unpublished observation). The ability to withstand stress in infected-NPC cells to interfere with the presentation of viral or
by EBV-infected cells may provide a growth advantage in vivo as tumour antigen to activate host innate and adaptive immunity. A
ischemia is common during exponential growth of tumor. genome-wide expression profiling in laser-captured, microdissect-
ed NPC tissues and normal nasopharyngeal epithelium showed
Contribution of stromal inflammation to growth of EBV-infected NPC strong correlation with the expression of EBV genes with inhibition
cells. A characteristic histopathological feature of NPC is the pres- of expression of multiple MHC class I HLA genes [95]. Similarly,
ence of exceptionally rich inflammatory stroma. EBV-infected down-regulation of locus-specific HLA Class I expression was also
NPC cells in patients are continuously confronted by inflammatory reported in EBV-associated gastric cancer [96]. The EBV proteins
cytokines present in the tumour stroma including TNF-a, TGF-b, could also directly impede the function of HLA molecules. The
IL6, IL8, etc. [86]. The effects of these inflammatory cytokines on Gly-Ala repeat (Gar) within the EBV EBNA1, which is expressed
the growth and malignant properties of EBV-infected NPC cells in all latent EBV-infected cells, inhibits MHC class 1 restricted anti-
are not fully understood. These inflammatory cytokines may sup- gen presentation [97]. The EBV BNLF2A could bind to the TAP
port the growth of EBV-infected NPC cells. Among these inflamma- transporter to inhibit the delivery of the antigenic peptide to HLA
tory cytokines, the IL6 was shown to support latent infection of molecules for presentation [98]. Furthermore, the EBV BILF1,
EBV through activation of STAT3 signaling [87]. STAT3 activation which function as a G protein-coupled receptor, increases endocy-
in EBV-infected cells upregulates the activity of the EBV Qp pro- tosis and lysosomal degradation of MHC class I molecules [99]. A
moter for EBNA1 transcription which is essential for latent infec- recent study also reported that the EBV BILF1 impedes presenta-
tion of EBV and maintenance of EBV episome in infected cells. tion of viral antigen and cytotoxic T cell recognition of the infected
The latent EBV gene, LMP1, could also upregulate transcription of cells [100]. The EBV BGLF5 also suppresses HLA antigen expression
IL6 in EBV-infected cells which activates STAT3 to upregulate as part of its action to shut off host immune function [101].
LMP1 transcription in EBV-infected NPC cells, constituting a posi- The EBV-encoded miRNAs have also been reported to partici-
tive feedback loop involving STAT3, LMP1 and IL6 to sustain activa- pate in the immune evasion of EBV-infected cells. The ebv-miR-
tion of STAT3 activation and latent EBV infection in NPC cells BART2-5p and ebv-miR-BART3 target MICB and IPO7 respectively
[87,88]. The action of STAT3 activation to enhance survival and [102]. The MICB is a cellular ligand for NKG2D which is a potent
malignant properties of human cancer cells is well-documented activating receptor presents on NK cells. The ebv-miR-BART2-5p
[89]. We recently reported that prolonged propagation of EBV-in- suppresses the expression of MICB in EBV-infected NPC cells pro-
fected immortalized nasopharyngeal epithelial cells (NP460) con- tecting them from recognition and killing by NK cells. The IPO7
ferred enhanced sensitivity to IL6 activation of STAT3 [90]. The (importin 7) is a nuclear importer receptor which contributes to
enhanced responsiveness of EBV-infected NP460 cells to IL6 activa- cytokine and early response gene expression in activated T cells.
tion of STAT3 was shown to be largely due to overexpression of IL6 The transportation of EBV-encoded miRNA from NPC tumor cells
receptor (IL-6R). IL-6R overexpression was detected in NPC speci- to neighboring TILs may impair their cytotoxic function through
mens. Overexpression of IL-6R also enhanced in vivo growth and inhibiting IPO7 expression.
tumorigenicity of C666-1, an EBV-positive NPC cell line, in immune Another study reported that EBV-infected cells commonly used
deficient animals. exosomes secretion to communicate with neighboring cells [103].
The IL8 (CXCL8) also supports spheroid growth and stemness Exosomes isolated from EBV-infected lymphoblasts (LCL) are en-
properties of EBV + ve NPC cells, C666-1 [91,92]. Overexpression riched in small RNAs and EBV-miRNAs which are delivered to
of IL8 is common in NPC tissues. Interestingly, the C666-1 cells also and internalized by monocyte-derived dendritic cells to repress
secrete IL8. An autocrine stimulating pathway may be involved to some of the EBV-targeted genes, including CXCL11/ITAC, which is
promote growth of EBV-infected NPC cells. an immunoregulatory gene down-regulated in primary EBV-asso-
Constitutive NF-jB signaling, which is proinflammatory in nat- ciated lymphomas [104]. NPC cells could produce exosomes con-
ure, is common in NPC and could be activated by either LMP1 taining high amount of galectin-9, a ligand of the membrane
expression or genetic alterations in EBV-positive NPCs. The major receptor Tim-1, to induce apoptosis in mature Th1 lymphocytes
NF-jB signals in NPC are p50/p50/BCL3 and p50/RELB [93]. By [105]. Recombinant galectin-9 could induce apoptosis in EBV-spe-
microarray analysis of c666-1 cells with p50 knockdown, we iden- cific CD4 + T cells and may play a role in the immune evasion of
tified multiple NF-jB downstream targets, including oncogenes EBV-infected NPC cells. Immune invasion may confer a distinct
(MYB, BCL2), chemokines and chemokine receptors (CXCL9, in vivo advantage of EBV-infected NPC cells.
CXCL10, CX3CL1 and CCL20) [94]. NPC is characterized by heavy
infiltration of tumour infiltrating lymphocytes (TILs). This has Contribution of latent and lytic EBV infection in NPC carcinogenesis
raised the possibility that chemokines induced by NF-jB activation Latent EBV infection is believed to be involved in tumorigenesis
in NPC cells may recruit TILs to the tumour microenvironment and as lytic reactivation of EBV infection in infected cells will lead to
interact with the NPC cells in various ways to support their growth cell death. In NPC, the lytic infection of EBV is generally believed
in vivo. The expression of CCL20 may also contribute to the escape to be abortive in nature without production of infectious viral par-
of EBV-infected NPC cells from host anti-viral immune response by ticles. Recent studies showed that both lytic and latent EBV genes
recruiting memory regulatory T-cells. The inflammatory stroma may contribute to the carcinogenesis of NPC.
could play important roles to promote growth and progression of The role of the latent infection of EBV in human carcinogenesis
NPC and may represent a druggable target for effective treatment has long been postulated. EBV readily established type II latency in
of the disease. NPC cells and expression of EBV genes, including EBERs, EBNA1,
LMP1, LMP2 and recently EBV-encoded miRNAs, have been shown
Evasion from immune surveillance. Immunosurveillance plays a vi- to be involved in tumorigenesis. Furthermore, latent infection of
tal role in elimination of tumour cells at early stage of EBV may drive epigenetic changes in host cell genome to promote
S.W. Tsao et al. / Oral Oncology 50 (2014) 330–338 335

tumorigenesis [106]. The LMP1 is considered as a classical viral factors, including consumption of salted fish and preserved food,
oncogene which activates multiple cell signaling, including NF- may contribute to the generation of genetic alterations in prema-
jB, MAPK and PI3K, to drive tumorigenesis [107]. A role of LMP1 lignant nasopharyngeal epithelial cells in high risk NPC popula-
in driving epigenetic changes has been reported. In NPC cells, the tions. EBV infection is consistently demonstrated in
LMP1 was shown to downregulate E-cadherin via activation of undifferentiated NPC. Genetic changes in premalignant nasopha-
DNA methyltransferases [108]. LMP1 could also directly activate ryngeal epithelial cells support EBV infection and its persistence
the DNMT1-P1 promoter via the JNK-AP-1 pathway [109]. In gas- after infection. The local stromal inflammatory cytokines have pro-
tric cancer, LMP2A was reported to induce DNMT1 overexpression found effects on growth of EBV-infected nasopharyngeal epithelial
through STAT3 activation and PTEN inactivation. The EBV EBNA1, cells. Multiple EBV genes, latent and lytic in nature, may contribute
which is ubiquitously expressed in EBV-associated human malig- to malignant transformation of premalignant nasopharyngeal epi-
nancies, could induce DNA damage through induction of reactive thelial cells. EBV infection and expression of viral genes including
oxygen species [110]. EBER-overexpressing EBV promotes in vivo the EBV-encoded miRNAs may enable the virally infected NPC cells
growth of infected NPC cells [111]. The BART transcript is abun- to evade immune surveillance and confer selective advantages for
dantly expressed in NPC [95]. The BART miRNAs encoded by EBV their growth in vivo. An intricate interplay of these factors is be-
are expressed at particularly high levels in EBV associated epithe- lieved to contribute to NPC pathogenesis (Fig. 1).
lial malignancies undergo type II latency [112]. The BART miRNAs With recent advances in the genomic studies, comprehensively
may have an important role in driving growth and malignant deciphering the gene-environment interactions may provide new
transformation in infected NPC cells. Multiples studies have re- opportunities for early diagnosis as well as therapeutic opportuni-
vealed that the EBV-encoded BART miRNAs target various cellular ties for NPC. In most of the endemic regions, screening of NPC is
and viral proteins involved in maintenance of viral latency, im- commonly limited to the patient’s family members. Identification
mune evasion, and cell survival in NPC [113]. of NPC-susceptibility loci and NPC-associated HLA using GWAS
While EBV infection in NPC cells is largely latent in nature, EBV analysis and next generation sequencing of a large panel of pa-
lytic genes, such as BZLF1, are invariably detected in clinical NPC tients with individual risk factor information will help to define
specimens [95]. The impact of lytic EBV infection in NPC carcino- the high-risk population for further diagnostic evolution and may
genesis is not fully understood. Lytic infection of EBV has been improve the chance of early cancer detection. Because of the con-
shown to be important in the development of EBV-induced lym- sistent association of EBV infection and NPC, EBV-specific anti-
phoma. A mutant EBV incapable to induce lytic infection is much body-based assays have been commonly used as screening and
less potent in induction of lymphoma in humanized mice reconsti- diagnostic markers in high-risk region. The development of real-
tuted with human fetal CD34 + hematopoietic stem cells [114]. time quantitative PCR analysis of EBV DNA in plasma has highly
Expression of lytic EBV genes may contribute to genomic instabil- improved the sensitivity and specificity for early detection of
ity in infected cells. The immediate-early protein, BZLF1 proteins, NPC. A recent study on a large cohort of healthy individuals has
which plays an integral role in activation of lytic EBV infection demonstrated that plasma DNA analysis is useful in screening for
could induce G2 and mitotic blocks in cells [115]. The BZLF1 pro- early NPC [120]. By targeting on the high risk population with de-
tein could also bind to chromosome during mitosis and may fined genetic factors will further enhance the effectiveness of early
sequester other BZLF-1 interacting proteins in cells to alter chro- detection of NPC. The success in the early screening program in en-
mosome compaction and chromatin structure [116]. The BZLF1 demic regions will greatly improve the current clinical manage-
also increases the level of acetylated histone and translocates ment of this deadly disease.
CBP to mitotic chromosomes. Another EBV lytic gene, BGLF4 could
induce early DNA condensation and abnormal mitosis [117].
Conflict of interest statement
Recurrent induction of lytic infection of EBV contributed to an en-
hanced tumorigenic phenotype in EBV-infected HONE1 cells [118].
None declared.
Prolonged DNA damage response in normal cells usually induces
apoptosis. Expression of multiple latent EBV genes, notably
LMP1, prevents apoptosis and enhances survival of EBV-infected Acknowledgements
cells [107]. By analogy to the classical two-stage carcinogenesis
model, EBV lytic infection may play an initiator role while EBV la- The authors acknowledged the generous support of the various
tent infection plays a role as promotor in carcinogenesis of NPC. funding sources from the Research Grant Council, Hong Kong: GRF
The inflammatory cytokines in the tumour stroma may play an (777809, 779810, 780911, 779312; 470708, 471709, 471610,
important role in regulating latent and lytic infection of EBV. The 471211, 470312); CRF (CUHK8/CRF/11R); AoE NPC Grant (AoE/M-
TGFb and its downstream mediators, could activate Zp promoter 06/08) and Theme-Based Research Scheme (T12-401/13-R). The
to activate transcription of the lytic EBV transactivator protein, authors would also like to acknowledge funding from the HMRF
BZLF1 protein, to initiate lytic infection of EBV [119]. In contrast, Grant, Hong Kong (12110942 and 13120872) and the CRCG Grant
the IL6 and possibly other cytokines induce STAT3 activation to from the University of Hong Kong.
support latent EBV infection. The interplay of stromal cytokines
in the regulation of latent and lytic infection of EBV may contribute References
to the tumorigenic transformation and progression of EBV-infected
nasopharyngeal epithelial cells. [1] Tsao SW, Lo KW, Huang DP. Nasopharyngeal carcinoma. In: Tselis AC, Jenson
H, editors. Epstein–Barr virus. New York: Taylor & Francis; 2006. p. 273–95.
[2] Raab-Traub N. Epstein–Barr virus in the pathogenesis of NPC. In: Robertson
ES, editor. Epstein–Barr virus Wymondham. Norfolk: Caister Academic Press;
Conclusion and future directions 2005. p. 71–92.
[3] Chang ET, Adami HO. The enigmatic epidemiology of nasopharyngeal
carcinoma. Cancer Epidemiol Biomarkers Prev 2006;15(10):1765–77.
The pathogenesis of NPC is a multi-stage event and involves [4] Lo KW, To KF, Huang DP. Focus on nasopharyngeal carcinoma. Cancer Cell
multiple etiological factors. The unique geographical distribution 2004;5(5):423–8.
and ethnicity of NPC incidence implicate strongly the involvement [5] Yu MC, Yuan JM. Epidemiology of nasopharyngeal carcinoma. Semin Cancer
Biol 2002;12(6):421–9.
of genetic susceptibility. A close association with HLA locus has [6] Lee AW, Foo W, Mang O, Sze WM, Chappell R, Lau WH, et al. Changing
been identified in high risk NPC families. The environmental epidemiology of nasopharyngeal carcinoma in Hong Kong over a 20-year
336 S.W. Tsao et al. / Oral Oncology 50 (2014) 330–338

period (1980–99): an encouraging reduction in both incidence and mortality. enzymes XRCC1 and hOGG1. Cancer Epidemiol Biomarkers Prev
Int J Cancer 2003;103(5):680–5. 2003;12(10):1100–4.
[7] Lee AW, Ng WT, Chan YH, Sze H, Chan C, Lam TH. The battle against [34] Nazar-Stewart V, Vaughan TL, Burt RD, Chen C, Berwick M, Swanson GM.
nasopharyngeal cancer. Radiother Oncol 2012;104(3):272–8. Glutathione S-transferase M1 and susceptibility to nasopharyngeal
[8] Tao Q, Chan AT. Nasopharyngeal carcinoma: molecular pathogenesis and carcinoma. Cancer Epidemiol Biomarkers Prev 1999;8(6):547–51.
therapeutic developments. Expert Rev Mol Med 2007;9(12):1–24. [35] Hildesheim A, Anderson LM, Chen CJ, Cheng YJ, Brinton LA, Daly AK, et al.
[9] Hsu C, Shen YC, Cheng CC, Hong RL, Chang CJ, Cheng AL. Difference in the CYP2E1 genetic polymorphisms and risk of nasopharyngeal carcinoma in
incidence trend of nasopharyngeal and oropharyngeal carcinomas in Taiwan: Taiwan. J Natl Cancer Inst 1997;89(16):1207–12.
implication from age-period-cohort analysis. Cancer Epidemiol Biomarkers [36] Chen Y, Chan SH. Polymorphism of T-cell receptor genes in nasopharyngeal
Prev 2006;15(5):856–61. carcinoma. Int J Cancer 1994;56(6):830–3.
[10] Wang H, Seow A, Lee HP. Trends in cancer incidence among Singapore [37] Zhou G, Zhai Y, Cui Y, Zhang X, Dong X, Yang H, et al. MDM2 promoter SNP309
Malays: a low-risk population. Ann Acad Med Singapore 2004;33(1): is associated with risk of occurrence and advanced lymph node metastasis of
57–62. nasopharyngeal carcinoma in Chinese population. Clin Cancer Res
[11] Jia WH, Huang QH, Liao J, Ye W, Shugart YY, Liu Q, et al. Trends in incidence 2007;13(9):2627–33.
and mortality of nasopharyngeal carcinoma over a 20–25 year period (1978/ [38] Zheng MZ, Qin HD, Yu XJ, Zhang RH, Chen LZ, Feng QS, et al. Haplotype of gene
1983–2002) in Sihui and Cangwu counties in southern China. BMC Cancer Nedd4 binding protein 2 associated with sporadic nasopharyngeal carcinoma
2006;6:178. in the southern Chinese population. J Transl Med 2007;5:36.
[12] Ali MY. Histology of the human nasopharyngeal mucosa. J Anat 1965;99(Pt [39] Wei YS, Kuang XH, Zhu YH, Liang WB, Yang ZH, Tai SH, et al. Interleukin-10
3):657–72. gene promoter polymorphisms and the risk of nasopharyngeal carcinoma.
[13] Gibb AG. Anatomy and development. In: Van Hasselt A, Gibb AG, editors. Tissue Antigens 2007;70(1):12–7.
Nasopharyngeal carcinoma. Hong Kong: Chinese University Press; 1999. p. [40] Ben Nasr H, Chahed K, Mestiri S, Bouaouina N, Snoussi K, Chouchane L.
31–60. Association of IL-8 (-251)T/A polymorphism with susceptibility to and
[14] Chan JKC, Pilch BZ, Kuo TT, Wenig BM, Lee AWM. Tumours of nasopharynx: aggressiveness of nasopharyngeal carcinoma. Hum Immunol
introduction. In: Barnes L, Eveson JW, Reichart P, Sidransky D, editors. WHO 2007;68(9):761–9.
classification of tumours: pathology and genetics of head and neck [41] Catarino RJ, Breda E, Coelho V, Pinto D, Sousa H, Lopes C, et al. Association of
tumours. Lyon: The International Agency for Research on Cancer (IARC); the A870G cyclin D1 gene polymorphism with genetic susceptibility to
2005. p. 83–84. nasopharyngeal carcinoma. Head Neck 2006;28(7):603–8.
[15] Chan JKC, Bray F, McCarron P, Foo W, Lee AWM, Yip T, et al. Nasopharyngeal [42] Yu MC, Ho JH, Lai SH, Henderson BE. Cantonese-style salted fish as a cause of
carcinoma. In: Barnes L, Eveson JW, Reichart P, Sidransky D, editors. WHO nasopharyngeal carcinoma: report of a case-control study in Hong Kong.
classification of tumours: pathology and genetics of head and neck Cancer Res 1986;46(2):956–61.
tumours. Lyon: The International Agency for Research on Cancer (IARC); [43] Yu MC, Mo CC, Chong WX, Yeh FS, Henderson BE. Preserved foods and
2005. p. 85–97. nasopharyngeal carcinoma: a case-control study in Guangxi. China Cancer
[16] Buell P. The effect of migration on the risk of nasopharyngeal cancer among Res 1988;48(7):1954–9.
Chinese. Cancer Res 1974;34(5):1189–91. [44] Yu MC, Huang TB, Henderson BE. Diet and nasopharyngeal carcinoma: a case-
[17] Jia WH, Feng BJ, Xu ZL, Zhang XS, Huang P, Huang LX, et al. Familial risk and control study in Guangzhou. China Int J Cancer 1989;43(6):1077–82.
clustering of nasopharyngeal carcinoma in Guangdong. China Cancer [45] Ning JP, Yu MC, Wang QS, Henderson BE. Consumption of salted fish and
2004;101(2):363–9. other risk factors for nasopharyngeal carcinoma (NPC) in Tianjin, a low-risk
[18] Bei JX, Jia WH, Zeng YX. Familial and large-scale case-control studies identify region for NPC in the People’s Republic of China. J Natl Cancer Inst
genes associated with nasopharyngeal carcinoma. Sem Cancer Biol 1990;82(4):291–6.
2012;22(2):96–106. [46] Aiyar A, Tyree C, Sugden B. The plasmid replicon of EBV consists of multiple
[19] Lu SJ, Day NE, Degos L, Lepage V, Wang PC, Chan SH, et al. Linkage of a cis-acting elements that facilitate DNA synthesis by the cell and a viral
nasopharyngeal carcinoma susceptibility locus to the HLA region. Nature maintenance element. EMBO J 1998;17(21):6394–403.
1990;346(6283):470–1. [47] Ho JHC. Genetic and environmental factors in nasopharyngeal carcinoma. In:
[20] Bei JX, Li Y, Jia WH, Feng BJ, Zhou G, Chen LZ, et al. A genome-wide association Nakahara W, Nishioka K, Hirayama T, Ito Y, editors. Recent advances in
study of nasopharyngeal carcinoma identifies three new susceptibility loci. human tumor virology and immunology Tokyo. University of Tokyo Press;
Nat Genet 2010;42(7):599–603. 1971. p. 275–95.
[21] Hildesheim A, Wang CP. Genetic predisposition factors and nasopharyngeal [48] Zheng X, Luo Y, Christensson B, Drettner B. Induction of nasal and
carcinoma risk: a review of epidemiological association studies, 2000–2011: nasopharyngeal tumours in Sprague–Dawley rats fed with Chinese salted
Rosetta Stone for NPC: genetics, viral infection, and other environmental fish. Acta Otolaryngol 1994;114(1):98–104.
factors. Sem Cancer Biol 2012;22(2):107–16. [49] Huang DP, Ho JH, Saw D, Teoh TB. Carcinoma of the nasal and paranasal
[22] Tse KP, Su WH, Chang KP, Tsang NM, Yu CJ, Tang P, et al. Genome-wide regions in rats fed Cantonese salted marine fish. IARC Sci Publ
association study reveals multiple nasopharyngeal carcinoma-associated loci 1978;20:315–28.
within the HLA region at chromosome 6p21.3. Am J Hum Genet [50] Zou XN, Lu SH, Liu B. Volatile N-nitrosamines and their precursors in Chinese
2009;85(2):194–203. salted fish–a possible etological factor for NPC in china. Int J Cancer
[23] Tse KP, Su WH, Yang ML, Cheng HY, Tsang NM, Chang KP, et al. A gender- 1994;59(2):155–8.
specific association of CNV at 6p21.3 with NPC susceptibility. Hum Mol Genet [51] Huang DP, Ho JH, Webb KS, Wood BJ, Gough TA. Volatile nitrosamines in salt-
2011;20(14):2889–96. preserved fish before and after cooking. Food Cosmet Toxicol
[24] Feng BJ, Huang W, Shugart YY, Lee MK, Zhang F, Xia JC, et al. Genome-wide 1981;19(2):167–71.
scan for familial nasopharyngeal carcinoma reveals evidence of linkage to [52] Zheng YM, Tuppin P, Hubert A, Jeannel D, Pan YJ, Zeng Y, et al. Environmental
chromosome 4. Nat Genet 2002;31(4):395–9. and dietary risk factors for nasopharyngeal carcinoma: a case-control study
[25] Xiong W, Zeng ZY, Xia JH, Xia K, Shen SR, Li XL, et al. A susceptibility locus at in Zangwu County, Guangxi. China Br J Cancer 1994;69(3):508–14.
chromosome 3p21 linked to familial nasopharyngeal carcinoma. Cancer Res [53] West S, Hildesheim A, Dosemeci M. Non-viral risk factors for nasopharyngeal
2004;64(6):1972–4. carcinoma in the Philippines: results from a case-control study. Int J Cancer
[26] Hu LF, Qiu QH, Fu SM, Sun D, Magnusson K, He B, et al. A genome-wide scan 1993;55(5):722–7.
suggests a susceptibility locus on 5p 13 for nasopharyngeal carcinoma. Eur J [54] Hildesheim A, West S, DeVeyra E, De Guzman MF, Jurado A, Jones C, et al.
Hum Genet 2008;16(3):343–9. Herbal medicine use, Epstein–Barr virus, and risk of nasopharyngeal
[27] He JF, Jia WH, Fan Q, Zhou XX, Qin HD, Shugart YY, et al. Genetic carcinoma. Cancer Res 1992;52(11):3048–51.
polymorphisms of TLR3 are associated with Nasopharyngeal carcinoma risk [55] Furukawa M, Komori T, Ishiguro H, Umeda R. Epstein–Barr virus early antigen
in Cantonese population. BMC Cancer 2007;7:194. induction in nasopharyngeal hybrid cells by Chinese medicinal herbs. Auris
[28] Zeng Z, Zhou Y, Zhang W, Li X, Xiong W, Liu H, et al. Family-based association Nasus Larynx 1986;13(2):101–5.
analysis validates chromosome 3p21 as a putative nasopharyngeal carcinoma [56] Zeng Y, Zhong JM, Mo YK, Miao XC. Epstein–Barr virus early antigen induction
susceptibility locus. Genet Med 2006;8(3):156–60. in Raji cells by Chinese medicinal herbs. Intervirology 1983;19(4):201–4.
[29] Song C, Chen LZ, Zhang RH, Yu XJ, Zeng YX. Functional variant in the 3’- [57] Jia WH, Qin HD. Non-viral environmental risk factors for nasopharyngeal
untranslated region of Toll-like receptor 4 is associated with nasopharyngeal carcinoma: a systematic review. Semin Cancer Biol 2012;22(2):117–26.
carcinoma risk. Cancer Biol Ther 2006;5(10):1285–91. [58] Hildesheim A, Dosemeci M, Chan CC, Chen CJ, Cheng YJ, Hsu MM, et al.
[30] Tiwawech D, Srivatanakul P, Karalak A, Ishida T. Cytochrome P450 2A6 Occupational exposure to wood, formaldehyde, and solvents and risk of
polymorphism in nasopharyngeal carcinoma. Cancer Lett nasopharyngeal carcinoma. Cancer Epidemiol Biomarkers Prev
2006;241(1):135–41. 2001;10(11):1145–53.
[31] Tiwawech D, Srivatanakul P, Karalak A, Ishida T. Glutathione S-transferase M1 [59] Epstein MA, Achong BG, Barr YM. Virus particles in cultured lymphoblasts
gene polymorphism in Thai nasopharyngeal carcinoma. Asian Pac J Cancer from Burkitt’s Lymphoma. Lancet 1964;1(7335):702–3.
Prev 2005;6(3):270–5. [60] Rickinson AB, Kieff ED. Epstein–Barr virus. In: Knipe DM, Howley PM, editors.
[32] Hirunsatit R, Kongruttanachok N, Shotelersuk K, Supiyaphun P, Voravud N, Fields virology. Philadelphia: Wolters Kluwer Health/Lippincott Williams &
Sakuntabhai A, et al. Polymeric immunoglobulin receptor polymorphisms Wilkins; 2007. p. 2656–700.
and risk of nasopharyngeal cancer. BMC Genet 2003;4:3. [61] Kieff ED, Rickinson AB. Epstein–Barr virus and its replication. In: Knipe DM,
[33] Cho EY, Hildesheim A, Chen CJ, Hsu MM, Chen IH, Mittl BF, et al. Howley PM, editors. Fields virology. Philadelphia: Wolters Kluwer Health/
Nasopharyngeal carcinoma and genetic polymorphisms of DNA repair Lippincott Williams & Wilkins; 2007. p. 2603–54.
S.W. Tsao et al. / Oral Oncology 50 (2014) 330–338 337

[62] Henle W, Henle G, Ho HC, Burtin P, Cachin Y, Clifford P, et al. Antibodies to [90] Zhang G, Tsang CM, Deng W, Yip YL, Lui VW, Wong SC, et al. Enhanced IL-6/IL-
Epstein–Barr virus in nasopharyngeal carcinoma, other head and neck 6R signaling promotes growth and malignant properties in EBV-infected
neoplasms, and control groups. J Natl Cancer Inst 1970;44(1):225–31. premalignant and cancerous nasopharyngeal epithelial cells. PLoS ONE
[63] Chien YC, Chen JY, Liu MY, Yang HI, Hsu MM, Chen CJ, et al. Serologic markers 2013;8(5):e62284.
of Epstein–Barr virus infection and nasopharyngeal carcinoma in Taiwanese [91] Lo MC, Yip TC, Ngan KC, Cheng WW, Law CK, Chan PS, et al. Role of MIF/
men. N Engl J Med 2001;345(26):1877–82. CXCL8/CXCR2 signaling in the growth of nasopharyngeal carcinoma tumor
[64] Xu J, Wan XB, Huang XF, Chan KC, Hong MH, Wang LH, et al. Serologic spheres. Cancer Lett 2013.
antienzyme rate of Epstein–Barr virus DNase-specific neutralizing antibody [92] Lun SW, Cheung ST, Cheung PF, To KF, Woo JK, Choy KW, et al. CD44+ cancer
segregates TNM classification in nasopharyngeal carcinoma. J Clin Oncol stem-like cells in EBV-associated nasopharyngeal carcinoma. PLoS ONE
2010;28(35):5202–9. 2012;7(12):e52426.
[65] Raab-Traub N. Epstein–Barr virus in the pathogenesis of NPC. Sem Cancer Biol [93] Thornburg NJ, Pathmanathan R, Raab-Traub N. Activation of nuclear factor-
2002;12(6):431–41. kappaB p50 homodimer/Bcl-3 complexes in nasopharyngeal carcinoma.
[66] Raab-Traub N, Flynn K. The structure of the termini of the Epstein–Barr virus Cancer Res 2003;63(23):8293–301.
as a marker of clonal cellular proliferation. Cell 1986;47(6):883–9. [94] Chung GT, Lou WP, Chow C, To KF, Choy KW, Leung AW, et al. Constitutive
[67] Pathmanathan R, Prasad U, Sadler R, Flynn K, Raab-Traub N. Clonal activation of distinct NF-kappaB signals in EBV-associated nasopharyngeal
proliferations of cells infected with Epstein–Barr virus in preinvasive carcinoma. J Pathol 2013;231(3):311–22.
lesions related to nasopharyngeal carcinoma. N Engl J Med [95] Sengupta S, den Boon JA, Chen IH, Newton MA, Dahl DB, Chen M, et al.
1995;333(11):693–8. Genome-wide expression profiling reveals EBV-associated inhibition of MHC
[68] Chan AS, To KF, Lo KW, Mak KF, Pak W, Chiu B, et al. High frequency of class I expression in nasopharyngeal carcinoma. Cancer Res
chromosome 3p deletion in histologically normal nasopharyngeal epithelia 2006;66(16):7999–8006.
from southern Chinese. Cancer Res 2000;60(19):5365–70. [96] Dutta N, Gupta A, Mazumder DN, Banerjee S. Down-regulation of locus-
[69] Chan AS, To KF, Lo KW, Ding M, Li X, Johnson P, et al. Frequent chromosome specific human lymphocyte antigen class I expression in Epstein–Barr virus-
9p losses in histologically normal nasopharyngeal epithelia from southern associated gastric cancer: implication for viral-induced immune evasion.
Chinese. Int J Cancer 2002;102(3):300–3. Cancer 2006;106(8):1685–93.
[70] Tsang CM, Yip YL, Lo KW, Deng W, To KF, Hau PM, et al. Cyclin D1 [97] Levitskaya J, Coram M, Levitsky V, Imreh S, Steigerwald-Mullen PM, Klein G,
overexpression supports stable EBV infection in nasopharyngeal epithelial et al. Inhibition of antigen processing by the internal repeat region of the
cells. Proc Natl Acad Sci USA 2012;109(50):E3473–3482. Epstein–Barr virus nuclear antigen-1. Nature 1995;375(6533):685–8.
[71] Hutt-Fletcher LM, Lake CM. Two Epstein–Barr virus glycoprotein complexes. [98] Wycisk AI, Lin J, Loch S, Hobohm K, Funke J, Wieneke R, et al. Epstein–Barr
In: Takada K, editor. Epstein–Barr virus and human cancer. Berlin; Hong viral BNLF2a protein hijacks the tail-anchored protein insertion machinery to
Kong: Springer; 2001. p. 51–64. block antigen processing by the transport complex TAP. J Biol Chem
[72] Jiang R, Gu X, Nathan CO, Hutt-Fletcher L. Laser-capture microdissection of 2011;286(48):41402–12.
oropharyngeal epithelium indicates restriction of Epstein–Barr virus [99] Zuo J, Currin A, Griffin BD, Shannon-Lowe C, Thomas WA, Ressing ME, et al.
receptor/CD21 mRNA to tonsil epithelial cells. J Oral Pathol Med The Epstein–Barr virus G-protein-coupled receptor contributes to immune
2008;37(10):626–33. evasion by targeting MHC class I molecules for degradation. PLoS Pathog
[73] Pegtel DM, Middeldorp J, Thorley-Lawson DA. Epstein–Barr virus infection in 2009;5(1):e1000255.
ex vivo tonsil epithelial cell cultures of asymptomatic carriers. J Virol [100] Griffin BD, Gram AM, Mulder A, Van Leeuwen D, Claas FH, Wang F, et al. EBV
2004;78(22):12613–24. BILF1 evolved to downregulate cell surface display of a wide range of HLA
[74] Frangou P, Buettner M, Niedobitek G. Epstein–Barr virus (EBV) infection in class I molecules through their cytoplasmic tail. J Immunol
epithelial cells in vivo: rare detection of EBV replication in tongue mucosa but 2013;190(4):1672–84.
not in salivary glands. J Infect Dis 2005;191(2):238–42. [101] Rowe M, Glaunsinger B, van Leeuwen D, Zuo J, Sweetman D, Ganem D, et al.
[75] Tsao SW, Tsang CM, Pang PS, Zhang G, Chen H, Lo KW. The biology of EBV Host shutoff during productive Epstein–Barr virus infection is mediated by
infection in human epithelial cells. Semin Cancer Biol 2012;22(2):137–43. BGLF5 and may contribute to immune evasion. Proc Natl Acad Sci USA
[76] Imai S, Nishikawa J, Takada K. Cell-to-cell contact as an efficient mode of 2007;104(9):3366–71.
Epstein–Barr virus infection of diverse human epithelial cells. J Virol [102] Skalsky RL, Corcoran DL, Gottwein E, Frank CL, Kang D, Hafner M, et al. The
1998;72(5):4371–8. viral and cellular microRNA targetome in lymphoblastoid cell lines. PLoS
[77] Shannon-Lowe CD, Neuhierl B, Baldwin G, Rickinson AB, Delecluse HJ. Resting Pathog 2012;8(1):e1002484.
B cells as a transfer vehicle for Epstein–Barr virus infection of epithelial cells. [103] Meckes Jr DG, Shair KH, Marquitz AR, Kung CP, Edwards RH, Raab-Traub N.
Proc Natl Acad Sci USA 2006;103(18):7065–70. Human tumor virus utilizes exosomes for intercellular communication. Proc
[78] Tugizov SM, Berline JW, Palefsky JM. Epstein–Barr virus infection of polarized Natl Acad Sci USA 2010;107(47):20370–5.
tongue and nasopharyngeal epithelial cells. Nat Med 2003;9(3):307–14. [104] Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van Eijndhoven MA,
[79] Lin CT, Kao HJ, Lin JL, Chan WY, Wu HC, Liang ST. Response of nasopharyngeal Hopmans ES, Lindenberg JL, et al. Functional delivery of viral miRNAs via
carcinoma cells to Epstein–Barr virus infection in vitro. Lab Invest exosomes. Proc Natl Acad Sci USA 2010;107(14):6328–33.
2000;80(8):1149–60. [105] Klibi J, Niki T, Riedel A, Pioche-Durieu C, Souquere S, Rubinstein E, et al. Blood
[80] Lin CT, Lin CR, Tan GK, Chen W, Dee AN, Chan WY. The mechanism of Epstein– diffusion and Th1-suppressive effects of galectin-9-containing exosomes
Barr virus infection in nasopharyngeal carcinoma cells. Am J Pathol released by Epstein–Barr virus-infected nasopharyngeal carcinoma cells.
1997;150(5):1745–56. Blood 2009;113(9):1957–66.
[81] Feederle R, Neuhierl B, Bannert H, Geletneky K, Shannon-Lowe C, Delecluse [106] Kaneda A, Matsusaka K, Aburatani H, Fukayama M. Epstein–Barr virus
HJ. Epstein–Barr virus B95.8 produced in 293 cells shows marked tropism for infection as an epigenetic driver of tumorigenesis. Cancer Res
differentiated primary epithelial cells and reveals interindividual variation in 2012;72(14):3445–50.
susceptibility to viral infection. Int J Cancer 2007;121(3):588–94. [107] Dawson CW, Port RJ, Young LS. The role of the EBV-encoded latent membrane
[82] Lu JH, Tang YL, Yu HB, Zhou JH, Fu CY, Zeng X, et al. Epstein–Barr virus proteins LMP1 and LMP2 in the pathogenesis of nasopharyngeal carcinoma
facilitates the malignant potential of immortalized epithelial cells: from (NPC). Semin Cancer Biol 2012;22(2):144–53.
latent genome to viral production and maintenance. Lab Invest [108] Tsai CN, Tsai CL, Tse KP, Chang HY, Chang YS. The Epstein–Barr virus
2010;90(2):196–209. oncogene product, latent membrane protein 1, induces the downregulation
[83] Tsang CM, Zhang G, Seto E, Takada K, Deng W, Yip YL, et al. Epstein–Barr virus of E-cadherin gene expression via activation of DNA methyltransferases. Proc
infection in immortalized nasopharyngeal epithelial cells: regulation of Natl Acad Sci USA 2002;99(15):10084–9.
infection and phenotypic characterization. Int J Cancer 2010;127(7): [109] Tsai CL, Li HP, Lu YJ, Hsueh C, Liang Y, Chen CL, et al. Activation of DNA
1570–83. methyltransferase 1 by EBV LMP1 Involves c-Jun NH(2)-terminal kinase
[84] Lo AK, Liu Y, Wang XH, Huang DP, Yuen PW, Wong YC, et al. Alterations of signaling. Cancer Res 2006;66(24):11668–76.
biologic properties and gene expression in nasopharyngeal epithelial cells by [110] Frappier L. Role of EBNA1 in NPC tumourigenesis. Semin Cancer Biol
the Epstein–Barr virus-encoded latent membrane protein 1. Lab Invest 2012;22(2):154–61.
2003;83(5):697–709. [111] Iwakiri D, Sheen TS, Chen JY, Huang DP, Takada K. Epstein–Barr virus-
[85] Lo AK, Lo KW, Ko CW, Young LS, Dawson CW. Inhibition of the LKB1-AMPK encoded small RNA induces insulin-like growth factor 1 and supports growth
pathway by the Epstein–Barr virus-encoded LMP1 promotes proliferation and of nasopharyngeal carcinoma-derived cell lines. Oncogene
transformation of human nasopharyngeal epithelial cells. J Pathol 2013. 2005;24(10):1767–73.
[86] Huang YT, Sheen TS, Chen CL, Lu J, Chang Y, Chen JY, et al. Profile of cytokine [112] Marquitz AR, Raab-Traub N. The role of miRNAs and EBV BARTs in NPC.
expression in nasopharyngeal carcinomas: a distinct expression of Semin Cancer Biol 2012;22(2):166–72.
interleukin 1 in tumor and CD4+ T cells. Cancer Res 1999;59(7):1599–605. [113] Lo AK, Dawson CW, Jin DY, Lo KW. The pathological roles of BART miRNAs in
[87] Chen H, Hutt-Fletcher L, Cao L, Hayward SD. A positive autoregulatory loop of nasopharyngeal carcinoma. J Pathol 2012;227(4):392–403.
LMP1 expression and STAT activation in epithelial cells latently infected with [114] Ma SD, Hegde S, Young KH, Sullivan R, Rajesh D, Zhou Y, et al. A new model of
Epstein–Barr virus. J Virol 2003;77(7):4139–48. Epstein–Barr virus infection reveals an important role for early lytic viral
[88] Chen H, Lee JM, Zong Y, Borowitz M, Ng MH, Ambinder RF, et al. Linkage protein expression in the development of lymphomas. J Virol
between STAT regulation and Epstein–Barr virus gene expression in tumors. J 2011;85(1):165–77.
Virol 2001;75(6):2929–37. [115] Mauser A, Holley-Guthrie E, Simpson D, Kaufmann W, Kenney S. The Epstein–
[89] Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a Barr virus immediate-early protein BZLF1 induces both a G(2) and a mitotic
leading role for STAT3. Nat Rev Cancer 2009;9(11):798–809. block. J Virol 2002;76(19):10030–7.
338 S.W. Tsao et al. / Oral Oncology 50 (2014) 330–338

[116] Adamson AL. Epstein–Barr virus BZLF1 protein binds to mitotic progression of nasopharyngeal carcinoma cells. Int J Cancer
chromosomes. J Virol 2005;79(12):7899–904. 2009;124(9):2016–25.
[117] Lee CP, Chen JY, Wang JT, Kimura K, Takemoto A, Lu CC, et al. Epstein–Barr [119] Israel BF, Kenney SC. EBV lytic infection. In: Robertson ES, editor. Epstein–
virus BGLF4 kinase induces premature chromosome condensation through Barr virus Wymondham. Norfolk: Caister Academic Press; 2005. p. 571–612.
activation of condensin and topoisomerase II. J Virol 2007;81(10):5166–80. [120] Chan KC, Hung EC, Woo JK, Chan PK, Leung SF, Lai FP, et al. Early detection of
[118] Fang CY, Lee CH, Wu CC, Chang YT, Yu SL, Chou SP, et al. Recurrent chemical nasopharyngeal carcinoma by plasma Epstein–Barr virus DNA analysis in a
reactivations of EBV promotes genome instability and enhances tumor surveillance program. Cancer 2013;119(10):1838–44.

You might also like