Skip to main content
Chinese Journal of Cancer logoLink to Chinese Journal of Cancer
. 2014 Nov;33(11):556–568. doi: 10.5732/cjc.014.10170

The interplay of host genetic factors and Epstein-Barr virus in the development of nasopharyngeal carcinoma

Maria Li Lung 1, Arthur Kwok Leung Cheung 1, Josephine Mun Yee Ko 1, Hong Lok Lung 1, Yue Cheng 1, Wei Dai 1
PMCID: PMC4244319  PMID: 25367335

Abstract

The interplay between host cell genetics and Epstein-Barr virus (EBV) infection contributes to the development of nasopharyngeal carcinoma (NPC). Understanding the host genetic and epigenetic alterations and the influence of EBV on cell signaling and host gene regulation will aid in understanding the molecular pathogenesis of NPC and provide useful biomarkers and targets for diagnosis and therapy. In this review, we provide an update of the oncogenes and tumor suppressor genes associated with NPC, as well as genes associated with NPC risk including those involved in carcinogen detoxification and DNA repair. We also describe the importance of host genetics that govern the human leukocyte antigen (HLA) complex and immune responses, and we describe the impact of EBV infection on host cell signaling changes and epigenetic regulation of gene expression. High-power genomic sequencing approaches are needed to elucidate the genetic basis for inherited susceptibility to NPC and to identify the genes and pathways driving its molecular pathogenesis.

Keywords: NPC genetics, Epstein-Barr virus, human leukocyte antigen, cell signaling, inflammation


Nasopharyngeal carcinoma (NPC) is an epithelial cancer whose etiology is associated with several factors, including infection with the ubiquitous human herpesvirus Epstein-Barr virus (EBV), host genetics, and environmental exposures. Although NPC is rare in most parts of the world, it is highly prevalent in southern Chinese populations. EBV plays an important role in the development of NPC, as most NPC tumors harbor this virus. This intimate relationship is attributed to the interaction of EBV with host cell genes for NPC development. It is now believed that EBV either selectively infects host cells with genetic alterations such as allelic loss[1] and aberrant cyclin D1 overexpression[2] or allows such cells to persist and eventually transform into cancerous cells.

Previous approaches for deciphering the molecular genetic basis for NPC included comparative genomic hybridization (CGH) and loss of heterozygosity (LOH) studies to determine copy number gains and losses, epigenetic studies to identify critically silenced candidate genes, and functional studies to identify critical regions and candidate genes associated with NPC tumorigenesis. Expression profiling and transcriptome analysis have also identified several candidate genes and signaling pathways of interest. These studies have revealed both oncogenes (Table 1) and tumor suppressor genes (Table 2) that are important in NPC development, including genes that are involved in transcriptional regulation; cell adhesion, growth, proliferation, migration, and invasion; cell cycle and apoptosis; angiogenesis; and epithelial-mesenchymal transition and metastasis.

Table 1. Oncogenes involved in nasopharyngeal carcinoma (NPC) development.

Gene name Tumor-associated functions
AKT (v-akt murine thymoma viral oncogene homolog 1) Induces metastasis[60]
BCAT1 (branched chain amino acid transaminase 1, cytosolic) Induces cell proliferation, migration, and invasion[61]
BCL2 (B-cell CLL/lymphoma 2) Inhibits apoptosis[62][64]
CCND1 (cyclin D1) Promotes cell cycle G1-S transition through regulation of pRb[65][67]
DeltaNp63/TP73L [tumor protein p73-like, p63 splicing variants lacking NH(2)-terminal transactivating domain] Regulates Notch signaling, cell proliferation, and cell death[68][70]
EGFR (epidermal growth factor receptor) Regulates cell signaling[71][75]
EIF4E (eukaryotic translation initiation factor 4E) Promotes cell cycle progression by up-regulation of c-Myc and MMP9[76]
EVI1 (ecotropic viral integration site 1) Regulates chromatin remodeling[77]
FGF3/Int-2 (fibroblast growth factor 3) Promotes cell growth and tumor growth and invasion[78]
ERBB2 (v-erb-b2 erythroblastic leukemia viral oncogene homologue 2) Controls cell proliferation and angiogenesis[74],[79]
HRAS (Harvey rat sarcoma viral oncogene homolog) Induces cell cycle progression, regulates cell motility, and plays a role in cell signaling[72],[80],[81]
ID1 (inhibitor of DNA binding 1; dominant negative helix-loop-helix protein) Regulates cell growth and senescence[82]
IL8 (interleukin-8) Promotes metastasis through activation of epithelial-mesenchymal transition and Akt[83]
MACC1 (metastasis-associated in colon cancer 1) Induces cell proliferation, migration, invasion, and colony formation[84]
MDM2 (MDM2 oncogene, E3 ubiquitin protein ligase) Interacts with p53, to regulate its ability to control cell cycle and apoptosis[85],[86]
MET (Met proto-oncogene) Regulates cell proliferation and is involved in cancer signaling pathways[87],[88]
MYC (v-myc avian myelocytomatosis viral oncogene homologue) Regulates transcription of BMI1; induces cell proliferation, apoptosis, cell cycle progression; increases the radiotolerance of cancer cells[64],[78],[80],[89],[90]
PIK3CA (phosphoinositide 3-kinase, catalytic, alpha polypeptide) Activates the activities of critical downstream cell signaling partners and enhances invasion[72],[91][93]
SATB1 (special AT-rich-binding protein 1) Decreases cell proliferation and resistance to apoptosis[94]
SP1 (SP1 transcription factor) Regulates transcription of BMI1[89]
TNFAIP3 (tumor necrosis factor, alpha-induced protein 3) Inhibits apoptosis and negatively regulates inflammatory response[95]

Table 2. Tumor suppressor genes involved in NPC development.

Gene name Tumor-associated functions
ADAMTS9 (A disintegrin and metalloproteinase with thrombospondin motifs 9) Inhibits angiogenesis by reduction of MMP9 and vascular endothelial growth factorA (VEGFA) expression[96],[97]
ADAMTS18 (A disintegrin and metalloproteinase with thrombospondin motifs 18) Activates diverse cell surface molecules, inhibits both anchorage-dependent and -independent growth[98]
BLU/ZMYND10 (zinc finger, MYND-type containing 10) Involves inhibition of angiogenesis, transcription factor stress response, and tumor suppression[99][101]
BRD7 (bromodomain containing 7) Regulates transcription and causes cell cycle arrest[102]
CDH1 (Cadherin 1, type 1, E-cadherin) Inhibits proliferation, invasion, and metastasis[103]
CDKN2A/p16 (cyclin-dependent kinase inhibitor 2A) Inhibits CDK4 kinase and causes cell cycle arrest[103]
CMTM3 (CKLF-like MARVEL transmembrane domain- containing 3) Involves cellular chemokine signaling[104]
CRIP2 (cysteine-rich intestine protein 2) Inhibits angiogenesis by transcriptional repression[105]
CRYAB (alpha B-crystallin) Suppresses tumorigenesis and epithelial-mesenchymal transition (EMT) by associating with adherens junction[106]
DLC1/ARHGAP7 (deleted in liver cancer 1) Involves cell cytoskeleton organization, activates GTPase, signal transduction, and cell adhesion, inhibits cell invasion[107]
DLEC1 (deleted in lung and esophageal cancer 1) Inhibits cell growth and invasiveness[108]
DUSP6 (dual specificity phosphatase 6) Suppresses cell proliferation, induces apoptosis, inhibits EMT by negatively regulating the activity of ERK[109]
FBLN2 (fibulin 2) Interacts with extracellular matrix (ECM) proteins; inhibits cell proliferation, migration, and invasion; suppresses angiogenesis[110]
FBLN3 (EGF-containing fibulin-like extracellular matrix protein 1) Suppresses migration and invasion of NPC cells and involves the regulation of Akt signaling pathways[111]
GADD45G (growth arrest and DNA damage-inducible, gamma) Involves DNA damage response, inhibits cell growth and colony formation[112]
IGFBP-6 (insulin-like growth for binding protein 6) Inhibits the proliferation, invasion, and metastasitic abilities; increases the apoptosis events of NPC cells; associates with the expression of EGR-1[113]
IRF8 (interferon regulatory factor 8) Affects host defense, cell growth, differentiation, immune regulation and inhibits clonogenicity[114]
LARS (leucyl-tRNA synthetase) Catalyzes ATP-dependent ligation of L-Leu to tRNA (leu) and is inactivated in NPC by both genetic and epigenetic mechanisms[115]
LTBP2 (latent transforming growth factor beta binding protein 2) Reduces focal adhesion and cell migration, suppresses angiogenesis[116]
MIPOL1 (mirror-image polydactyly 1) Arrests cell cycle transition[117]
MMP19 (matrix metallopeptidase 19) Breaks down ECM to affect cell proliferation, migration, and adhesion[118]
PCDH10 (protocadherin 10) Mediates cell-cell adhesion, induces apoptosis, and be involved in cell signaling[119]
PTPRG (protein tyrosine phosphatase receptor type G) Arrests cell cycle, involves cell-ECM interactions, dephosphorylates kinases[120]
RASSF1A [ras association (RalGDS/ AF-6) domain family 1] Involves cell cycle arrest, induces apoptosis, involves DNA repair, inhibits accumulation of cyclin D1[121]
THY1/CD90 (Thy-1 cell-surface antigen) Involves suppression of tumor formation, cell proliferation, and invasion[122],[123]
TSLC1/CADM1 (tumor suppressor in lung cancer 1/cell adhesion molecule 1) Inhibits cell growth and induces apoptosis[124],[125]
WIF1 (WNT inhibitory factor 1) Inhibits WNT proteins, involves protein-tyrosine kinase activity[126]
ZNF382 (KRAB zinc finger protein) Inhibits proliferation, induces apoptosis[127]

More recently, genome-wide association studies (GWAS) and single nucleotide polymorphism (SNP) analyses have identified candidate genes and aberrant pathways of importance in NPC. Multiple genetic regions were identified in familial and case-control studies (Table 3), indicating the possibility of multi-factorial risk factors and the combination of common low penetrance alleles having a role in NPC genetic risk.

Table 3. Familial and case-control genomic studies in NPC.

Study type Location Details Results
Familial Guangzhou 20 families
382 microsatellite markers covering 22 auto- somes with average marker density of 10 cM
Linkage to chromosome (chr) 4p15.1-q12 (14.21 cM)[128]
Familial Hunan 18 families
20 microsatellite markers from chr 4p15-q12 (5), chr 3p (8), chr 9p (7)
Linkage to chr 3p21.3-21.2 (13.6 cM)[129]
Familial Guangzhou 15 families
800 microsatellite markers covering 22 auto- somes with average marker density of 5 cM
Linkage to chr 5p13 (17 cM)[39]
Case-control Guangxi 350 NPC cases, 634 controls chr 4p not confirmed[130]
Study of chr 4p15.1-q12 region with 34 microsatellite markers
Case-control Malaysia 111 NPC cases, 260 controls
Genome-wide association study (GWAS) with 500,000 tag single nucleotide polymorphisms (SNPs)
Identified SNP in intron 3 of ITGA9 (integrin, α9) on 3p21 (40 kb)[15]
Case-control Taiwan 277 NPC cases, 285 controls
GWAS with 480,000 SNPs; biological role for GABBR1 in NPC
Linkage to HLA region at 6p21.3 in HLA-A & F/GABBR1 (GABA receptor 1) genes[14]
Case-control Guangzhou, Guangxi 1,583 NPC cases, 1,894 controls
GWAS with 464,000 autosomal SNPs
Linkage to 13q12 (TNFRSF19, TNF receptor superfamily 19), 3q26 (MDS1-EVI1), 9p21 (CDKN2A-CDKN2B) and reconfirm HLA on chr 6[13]
Case-control Hong Kong 360 NPC cases, 360 controls
MassArray Sequenom SNP study with 233 SNPs confined to 6p
GABBR1, HLA-A, and HCG9 were highly associated with NPC in single- marker association studies; microdeletions in GABBR1 and NEDD9 (neural precursor cell expressed developmentally down-regulated 9) were detected[131]

Understanding the molecular pathogenesis of EBV infection and host gene aberrations will aid in our strategies for diagnostics and treatment of NPC. Human leukocyte antigen (HLA) affects host responses to EBV infection through viral antigen presentation. Inhibition of HLA expression may facilitate tumor cell evasion of the normal host immunosurveillance. Several studies support the link between the HLA complex, genetic susceptibility to NPC, and immune response to EBV (Table 4). Environmental exposure to carcinogens also plays a role in NPC development. Many studies have examined the link between NPC risk and carcinogen metabolism (Table 5) or DNA repair (Table 6).

Table 4. Genetic risk for NPC: HLA and immune responses to Epstein-Barr virus (EBV).

Gene/function Study
Human leukocyte antigen (HLA) Linkage analysis studies in Hong Kong and Singapore show HLA association with NPC[18]
HLA-A2-B46 haplotype is associated with NPC in Taiwan[16]
RFLP study in Tunisia shows HLA-G (facilitates escape from cancer immunosurveillance); Ile 110 allele is less frequent among patients with lymph node involvement and more severe tumor stage, and deletion of C in codon 130 was associated with decreased NPC-free disease and survival[132]
T-cell receptor (TCR) and Toll-like receptor (TLR) in EBV infection and immune response PCR-RFLP analysis of TCR gene in Singaporean study shows TCR polymorphism is associated with decreased NPC risk, particularly in patients with HLA B46[133]
PCR and direct sequencing of TLR polymorphism in Guangzhou study shows TLR3 polymorphism is related to NPC susceptibility but the effect is modest[134]
TLR4 SNP may modulate immune response to EBV and predispose to NPC[135]
Host cell immune response to EBV Microarray profiling of tumor and normal. EBV latent genes were confirmed to strongly associate with suppression of MHC class I HLA gene[4]
DC-SIGN promoter SNP analysis in a Cantonese population shows two SNPs on DC-SIGN promoter are associated with high risk for NPC[136]
CTLA-4 polymorphism analysis in a Hangzhou study shows that CTLA-4 SNP is highly associated with NPC susceptibility[137]
TNFα and HSP70-2 polymorphisms in a Tunisian study show the HSP70-2 genotype is associated with increased NPC risk[138]
PCR-RFLP analysis in a Taiwan study shows that p21WAF/CIP1 and TNFα polymorphisms have no association with NPC; comparison between smokers and non-smokers shows the association of environmental factor with the p21 in NPC[139]
TNFα polymorphism in Portuguese study shows NPC risk increased in undifferentiated NPC[140]
Miscellaneous Genotyping in a Guangzhou study shows an association of EBV-positive serology and genetic factors represented by tag SNPs in 35 genes in homologous recombination repair involved in DNA repair among healthy individuals[141]

PCR-RFLP, polymerase chain reaction-restriction fragment length polymorphism.

Table 5. Genetic risk for NPC: carcinogen metabolism.

Enzyme(s) Function Study Results
CYP2E1 Carcinogen metabolism 50 NPC cases and 50 controls & 364 NPC cases and 320 controls in Taiwan were analyzed by PCR-RFLP Increased NPC risk for homozygous variant genotype[142],[143]
Carcinogen metabolism 2,499 subjects from 546 NPC families were genotyped547 NPC cases and 755 controls in Guangzhou Association of SNP and increased NPC risk for individuals <46 years and with smoking history[144]
CYP2A6 Carcinogen metabolism 74 NPC cases and 137 controls in Thailand were analyzed by PCR-RFLP 5-fold increase in NPC risk with mutant allele[145]
CYP2A13 Carcinogen metabolism The CYP2A13 gen from 45 NPC patients in Guangzhou were PCR-amplified and sequenced Identified novel SNPs, but no correlation between SNPs and NPC risk[146]
CYPE2B6, CYPE2E1, PRKDC, PCNA, CHEK2, NQ01 DNA repair, nitrosamine metabolism 31 NPC cases and 10 controls in Taiwan compared in a microarray targeting biological pathways for carcinogen metabolism, DNA repair, and chromosomal regions of interest Differential expression in genes for DNA repair, nitrosamine metabolism, chromosomes 4p15-4q12 and 14q32[147]
CYP2E1, GSTP1, NQO1, MPO Carcinogen metabolism 358 NPC cases and 629 controls in Guangzhou and Guangxi studied with Taqman genotyping and Tag SNPs No significant difference between cases and controls[148]
GSTM1 (glutathione S-transferase M1) Carcinogen metabolism 83 NPC cases and 114 controls in the US No association with NPC risk, but absence of GSTM1 is associated with moderately increased NPC risk[149]
GSTM1, GSTT1 (glutathione S-trans-ferase theta-1) Carcinogen metabolism 350 NPC cases and 622 controls in Beijing studied with multiplex PCR No significant association with NPC risk, but males with double null genotype had increased NPC risk[150]
GSTM1, GSTT1 Carcinogen metabolism Meta-analysis of 85 published papers and selected 8 case-control studies of NPC GSTM1 deletion is a risk factor for NPC; no association of GSTT1 with NPC risk[151]

Abbreviation as in Table 4.

Table 6. Genetic risk for NPC: DNA repair.

Enzyme(s) Function Study Results
XRCC1 (X-ray repair complementing defective repair in Chinese hamster cells 1), hOGG1 (human 8-oxoG DNA glycosylase) (CYP2E1) DNA repair 334 NPC cases and 283 controls in Taiwan were studied with PCR-RFLP Increased odds ratio (OR ) with multiple putative high-risk genotypes. Carriers with 1 putative high-risk genotype had OR=3; with 2, OR=4.3; and with 3, OR=25.[20]
XRCC1 XPD (xeroderma pigmentosum group D or ERCC2) DNA repair 462 NPC cases and 511 controls in Guangzhou were studied with PCR-RFLP XRCC1 variant genotype associated with decreased NPC risk, especially among males and smokers[152]
153 NPC cases and 168 controls in Sichuan were studied with PCR-RFLP Increased NPC risk with XRCC1 and borderline decrease in NPC risk with XPD; if both alleles are involved, then increase in NPC risk. No association with XRCC3[153]
ERCC1 (excision repair cross- complementing rodent repair deficiency, complementation group overlapping antisense sequence) DNA repair 267 NPC cases and 304 controls in Sichuan were studied with PCR-RFLP ERCC1 polymorphism associated with NPC risk[154]
ERCC1 genotyping in 42 patients with NPC in Hong Kong treated with gemcitabine and oxaliplatin No associations between survival or response rate and ERCC1 genotype[155]
RAD51L1 (RAD51 paralog B), BRCA2 (breast cancer 2, early onset), TP53BP1 (tumor protein p53-binding protein 1) DNA repair Discovery stage: 755 NPC cases and 755 controls in Guangzhou were studied by GoldenGate genotyping platform to investigate 676 tagging SNPs for 88 DNA repair genes. Individuals with inherited defects in DNA repair genes have increased NPC risk; RAD51L1 was the only gene validated[156]
Validation stage: 1,568 NPC cases and 1,297 controls were analyzed by Sequenom DNA MassARRAY to validate 11 SNPs
N4BP2 (Nedd4-binding protein 2) DNA repair 531 NPC cases and 480 controls in Guangzhou studied with PCR sequencing Identified 3 novel SNPs associated with N4BP2 in NPC susceptibility 4p15 locus; two haplotypes were associated with NPC[157]

Abbreviation as in Table 4.

NPC tumors characteristically contain large numbers of infiltrating lymphocytes, and EBV-induced inflammation is often associated with STAT3 activation. Latent membrane protein 1 (LMP1) is reported to sensitize NPC cells to genotoxic drugs. Nuclear factor kappaB (NF-κB) signaling is associated with NPC tumor formation[3]. In this review, we discuss the associations of NPC with HLA and inflammation and with aberrant cell signaling. We also describe in brief next-generation sequencing (NGS) approaches to better understand the associations between host genetics and EBV infection.

HLA and Inflammation

Host immune responses are important in determining the consequence of viral infection–related cancers. EBV plays an integral role in tissue inflammation and NPC development. The tumorigenic potential of viral infections is associated with their carriage of genes associated with cell transformation and their ability to induce chronic inflammation. The HLA system plays a central role in viral antigen presentation, which is key to determine the outcome of the host immune response to this lifelong viral infection. HLA genes are believed to play a role in NPC development because they have a functional impact on the innate and adaptive immune responses against the viral etiologic agent, EBV. NPC cells expressing specific EBV proteins, which are processed and the antigen presented in association with HLA class I alleles, may be recognized by EBV-specific CD8+ cytotoxic T cells. Some evidence supports the hypothesis that EBV may down-regulate the expression of HLA alleles and result in immune escape of NPC cancer cells by decreasing the recognition of EBV-expressing cancer cells[4][6].

The genetic association of the major histocompatibility complex (MHC) region, in which HLA resides, is validated by a catalog of GWAS studies with numerous diseases and conditions including inflammatory, autoimmune, and infectious diseases; cancer; drug-induced hypersensitivity; and neuropsychiatric disease[7] . Historically, the first association of HLA alleles with NPC was reported in 1974[8]. For EBV-associated tumors such as NPC, previous candidate gene studies have shown a strong linkage between HLA associations and NPC risk[8][12]. These findings have been verified in more recent, large-scale GWAS in different populations[13][15]. Linkage analysis and several recent SNP studies indicate a strong association between HLA-associated genes and NPC risk[16][19]. The association of the HLA class I genes and risk of NPC development was reported in different ethnic Chinese populations including Singaporean Chinese[12],[20], Taiwanese[11],[16],[21],[22], and southern Chinese[9],[23]. The HLA class I alleles HLA-A2 and HLA-B46 are most consistently associated with increased susceptibility for NPC in high-risk regions and among the Chinese population. On the other hand, HLA alleles HLA-A11, HLA-B13, HLA-B27, and HLA-A31 confer protective effects for NPC risk in high-risk areas. More details and comprehensive information regarding HLA associations with NPC are provided in several well-written reviews on NPC genetic predisposition[24][26]. However, the distribution of frequencies of HLA alleles or haplotypes varies among high-/intermediate-/low-risk regions and resulted in different, sometimes inconsistent, and even opposite associated alleles in different geographic areas. Such an example is HLA-A2, which is associated with lower NPC risk in low incidence areas[27],[28]. This phenomenon may be due to an alternative hypothesis that HLA may only be a genetic marker, which is in close linkage with another NPC predisposition locus. The HLA region is highly polymorphic with enormous sequence diversity and gene density, and the genes are in extensive linkage disequilibrium. Thus, this complexity hinders the hunt for causal variants for NPC development. Standard molecular tools such as polymerase chain reaction-based methods for genotyping, including MassArray Sequenom, Taqman assay, and the GoldenGate Assay, have limited capacity, and probe designs are restricted by this difficulty for the HLA region. As most studies focus on 4-digit coding variation, the role of some functional non-coding variants within the HLA region may not have been addressed. There is a need for studying the role of non-coding variants of HLA and non-HLA genes and for identifying novel rare and common variants in NPC genetic susceptibility by applying deep resequencing approaches with the technological advances in NGS. To elucidate functional mechanisms and disease pathogenesis, it is crucial to identify the causal variants associated with NPC. HLA undoubtedly plays a substantial role in genetic predisposition for NPC development. Findings from previous studies that examined the specific EBV epitopes from LMP1 and LMP2 recognized by T cells in association with specific HLA alleles support the hypothesis that HLA alleles associated with NPC risk affect the processing and antigen presentation[6],[29][31]. Specific HLA class I alleles and amino acid variants (HLA-A*11:01 allele, HLA-B*13:01, and B*55:02) in the HLA class I peptide-binding groove were observed to confer higher NPC risk[10]. However, additional studies are needed to clarify how the HLA class I antigen recognition groove and the EBV epitopes interact. HLA class I alleles possess another function related to innate immunity which modulates the natural killer (NK) cells that kill cancer cells. HLA class I molecules induce the activation of killer-immunoglobulin-like receptors (KIRs), which in turn inhibit the function of NK cells. NPC risk is higher for EBV-seropositive individuals carrying at least five activating KIRs compared with those without activating KIRs[32]. By revealing the detailed insights into the HLA functional variants for NPC, potential novel therapeutic options will be developed.

EBV Latency Genes Induce Host Cell Signaling Changes

EBV usually undergoes different types of latency in various cell types. After EBV infection in the epithelial cell, EBV usually enters type II latency and expresses a specific panel of latent proteins including the LMP1, LMP2, and EBNA1. Although EBV infection is frequently observed in NPC, the contribution of these oncogenic latent proteins to the pathogenesis of NPC is not as clear as that in the B-cell transformation.

Increasing evidence supports the involvement of LMP1 in altering host oncogenic signaling including the NF-κB, Akt, JNK1/c-jun, and MEK/ERK-MAPK pathways in different cancers such as NPC. The C-terminal activating region 2 (CTAR2) of LMP1 is important for activation of the NF-κB signaling via the host cell proteins TRAF6 and TAK1[33],[34]. LMP1 induces NF-κB–associated transcriptional activation to drive the expression of TNFAIP2 and, thus, promotes migration[35]. On the other hand, LMP1 also showed inhibitory effects on tumor suppressive signaling pathways, including the LKB1-AMPK pathway. The LMP1 CTAR1 can activate the MEK/ERK-MAPK signaling pathway, resulting in phosphorylation of LKB1 and subsequent suppression of AMPK activity[36]. LMP1 was also found to induce the epithelial-mesenchymal transition (EMT) via the regulation of Twist, a master transcriptional regulator in embryogenesis and metastasis, through the NF-κB signaling pathway. The induction of EMT by Twist contributes to increased cell motility and invasiveness and, thus, resulted in more metastatic characteristics in NPC[37]. Angiogenesis is another important event for cancer development, and EBV also plays a role in regulating this process. LMP1 was reported to induce the expression of the principal pro-angiogenic factor vascular endothelial growth factor (VEGF) via the activation of JAK/STAT and MAPK/ERK signaling pathways[38]. In addition to LMP1, the EBV latent protein EBNA1 can increase the activity of the AP1 transcription factor and induce AP1-mediated up-regulation of IL8, VEGF, and hypoxia-inducible factor-1α to enhance angiogenesis in vitro[39].

Although LMP2A plays a well-known, essential role in maintaining EBV latency in B cells, its role in epithelial cells is not well understood. Unlike LMP1, LMP2A is not normally regarded as a viral transforming gene and its expression is more consistently observed in NPC than LMP1. Indeed, the association of LMP2 with various oncogenic cell signaling pathways has been reported, suggesting that LMP2A may also participate in EBV-induced epithelial cell transformation[40]. For example, LMP2A can increase cell migration via the Akt signaling pathway by phosphorylating an inhibitory site on GSK3β, a Wnt signaling modulator downstream of Akt[41]. Indeed, LMP2A can activate the Akt signaling pathway in a PI3K-dependent manner and result in a PI3K-dependent nuclear translocation of β-catenin in the human foreskin keratinocytes[42].

EBV Impacts Stem Cell Signaling Pathways

Accumulating evidence indicates that understanding of cancer stem cell (CSC) behaviors holds great promise for the treatment of human cancers. However, CSC studies in NPC have been greatly hampered by the lack of suitable markers for investigation. Out of very few reports of the NPC CSC studies, the CD44+SOX2+ minor population was found to have stem-like properties in EBV-positive C666 NPC cells[43]. EBV seems to play an important role in regulating the stem-like characteristics, as the expression of the EBV latent proteins was associated with the activation of the Hedgehog (HH) signaling pathway and the expression of stemness-related markers and genes[44].

One important hallmark for both cancer and stem cells is their self-renewal capability. As yet, it is not fully understood how cells regulate their own proliferation and differentiation into various tissues and cells. Many signaling pathways have been linked to stem cell behaviors or self-renewal abilities. Among them, Wnt, HH, and Notch are prominent signaling pathways reported over the past decade. Evidence gathered to date indicates these pathways may have regulatory impacts on stem cell growth and differentiation. We recently found that Wnt signaling regulates self-renewal networks, cyclin D1 expression, p53 pathway, generation of stem-like cells, and growth abilities of NPC cells[45]. Because EBV has been regarded as a major cause of NPC, it is reasonable to speculate that EBV may directly affect both p53 and Wnt signaling during the development of NPC[46],[47].

EBV Induces Epigenetic Changes in NPC

Epigenetic modifications, including histone modification and promoter hypermethylation, are critical for NPC tumor development. The LMP1 protein was reported to induce up-regulation of DNA methyltransferase (DNMT1) expression[48]. DNMT1 is mainly responsible for the maintenance of DNA methylation[49]. DNMT1 expression was indeed induced by the LMP1 CTAR2 YYD domain via the JNK/AP1 signaling pathway. Thereafter, activated DNMT1 expression resulted in promoter hypermethylation of the key epithelial marker E-cadherin[48]. On the other hand, EBV is also associated with histone modification in NPC. LMP1 expression positively correlated with the degree of phosphorylation of the serine 10 residue in histone H3 (p-H3Ser10). As a result, this histone modification is associated with increased cell proliferation, foci formation, and AP1 activation in NPC[50]. In contrast, knockdown of histone H3 or overexpression of a dominant-negative mutant (H3S10A) reversed the above-mentioned phenotypes. This suggests an important role of EBV in regulating epigenetic changes in cancer-related genes of the host cells in NPC and highlights important interactions of host and viral genes.

NGS Approaches to Elucidating the Molecular Genetics of NPC

GWAS studies using SNP arrays show the association of HLA subtypes with NPC susceptibility and identified several additional susceptibility loci[10],[13][15]. However, these studies are often limited by the loci on the array, with most being noncoding or far away from genes. Hence, these loci are not immediately informative and are difficult to be studied experimentally. Moreover, GWAS studies often target common variants and miss the rare ones that might underlie cancer genetics[51]. Therefore, additional GWAS are needed to understand the genetic basis of NPC. Recent advances in NGS approaches have allowed NPC researchers to systematically sequence expressed genes (“transcriptome”), known exons (“exomes”), and complete genomes in NPC, as well as the EBV genome, to decipher the regulatory network between EBV and host[52]. Recently, Szeto et al.[53] used RNASeq to characterize the sequence variants and the mRNA-microRNA regulatory network in NPC cancer cell lines. Liu et al.[54] applied the NGS system to assemble the EBV genome using samples from patients with NPC in Guangdong province. An accumulation of such studies would help to elucidate NPC pathogenesis.

The tremendous data generated from NGS approaches provides a statistical and computational challenge. Although bioinformatics tools for computational analysis have developed rapidly[55][57], the choice of analysis is not straightforward and depends on the specific aims and study design. Thus, data analysis and interpretation remain the bottleneck of cancer genomic studies due to the complexities of NGS data and the cancer genome. Another challenge in NPC genomic study is significant heterogeneity and lymphocyte infiltration in the specimens, which might mask the true biological events in tumor cells[58]. To minimize false-negative errors, the International Cancer Genome Consortium (ICGC) guidelines suggest that the tumor cell content of a sample be at least 60%–80%. Meanwhile, increasing redundant coverage in sequencing can help compensate for low tumor purity[57]. It is also a great challenge for bioinformaticians to develop tools that allow sensitive detection of genomic changes in impure and heterogeneous cancer samples[59]. To minimize false-positives, verification resequencing is necessary to estimate the technology-relevant errors, and it is important for studies to report a verification rate. As proposed by ICGC, at least 95% of the mutations identified in each sample should be real.

To make biological sense of the findings from NGS data will require computational, biological, and clinical analyses to link the biological pathways and the functional relevance of the molecular alterations to NPC, and to evaluate the associations of genomic changes with NPC diagnosis, prognosis, and treatment strategies. To tackle the fundamental basis for NPC, basic scientists in the area of bioinformatics, EBV, and NPC must collaborate with oncologists who can provide clinical information and translate our findings to the clinic. Furthermore, establishment of a NPC tissue bank is necessary to provide key resources for NGS projects. We have established a Hong Kong-wide Center for NPC Research (www.cnpcr.hku.hk) that aims to use genome-wide NGS approaches to elucidate the genetic basis for NPC susceptibility and to identify the genes and the biological pathways that underlie aggressive, recurrent NPC.

Conclusions

Clarifying the roles of host and EBV genetics in NPC development is expected to enhance our understanding of NPC pathogenesis and to provide improved biomarkers for detection and novel targets for therapeutic intervention. Targeted therapeutics will enhance survival of NPC patients with metastatic disease. Therefore, elucidating the interactions between EBV and host genes is expected to improved strategies for the clinical management of this deadly cancer.

Acknowledgments

This work was supported by the Research Grants Council Area of Excellence grant (AoE/M-06/08) to MLL.

References

  • 1.Lo KW, Huang DP. Genetic and epigenetic changes in naso-pharyngeal carcinoma. Semin Cancer Biol. 2002;12:451–462. doi: 10.1016/s1044579x02000883. [DOI] [PubMed] [Google Scholar]
  • 2.Tsang CM, Yip YL, Lo KW, et al. Cyclin D1 overexpression supports stable EBV infection in nasopharyngeal epithelial cells. Proc Natl Acad Sci U S A. 2012;109:E3473–3482. doi: 10.1073/pnas.1202637109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Chung GT, Lou WP, Chow C, et al. Constitutive activation of distinct NF-kappaB signals in EBV-associated nasopharyngeal carcinoma. J Pathol. 2013;231:311–322. doi: 10.1002/path.4239. [DOI] [PubMed] [Google Scholar]
  • 4.Sengupta S, den Boon JA, Chen IH, et al. Genome-wide expression profiling reveals EBV-associated inhibition of MHC class I expression in nasopharyngeal carcinoma. Cancer Res. 2006;66:7999–8006. doi: 10.1158/0008-5472.CAN-05-4399. [DOI] [PubMed] [Google Scholar]
  • 5.Ogino T, Moriai S, Ishida Y, et al. Association of immunoescape mechanisms with Epstein-Barr virus infection in nasopharyngeal carcinoma. Int J Cancer. 2007;120:2401–2410. doi: 10.1002/ijc.22334. [DOI] [PubMed] [Google Scholar]
  • 6.Lin HJ, Cherng JM, Hung MS, et al. Functional assays of HLA A2-restricted epitope variant of latent membrane protein 1 (LMP-1) of Epstein-Barr virus in nasopharyngeal carcinoma of Southern China and Taiwan. J Biomed Sci. 2005;12:925–936. doi: 10.1007/s11373-005-9017-y. [DOI] [PubMed] [Google Scholar]
  • 7.de Bakker PI, Raychaudhuri S. Interrogating the major histo-compatibility complex with high-throughput genomics. Hum Mol Genet. 2012;21:R29–36. doi: 10.1093/hmg/dds384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Simons MJ, Day NE, Wee GB, et al. Nasopharyngeal carcinoma V: immunogenetic studies of Southeast Asian ethnic groups with high and low risk for the tumor. Cancer Res. 1974;34:1192–1195. [PubMed] [Google Scholar]
  • 9.Tang M, Zeng Y, Poisson A, et al. Haplotype-dependent HLA susceptibility to nasopharyngeal carcinoma in a Southern Chinese population. Genes Immun. 2010;11:334–342. doi: 10.1038/gene.2009.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Tang M, Lautenberger JA, Gao X, et al. The principal genetic determinants for nasopharyngeal carcinoma in China involve the HLA class I antigen recognition groove. PLoS Genet. 2012;8:e1003103. doi: 10.1371/journal.pgen.1003103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hildesheim A, Apple RJ, Chen CJ, et al. Association of HLA class I and II alleles and extended haplotypes with nasopharyngeal carcinoma in Taiwan. J Natl Cancer Inst. 2002;94:1780–1789. doi: 10.1093/jnci/94.23.1780. [DOI] [PubMed] [Google Scholar]
  • 12.Chan SH, Day NE, Kunaratnam N, et al. HLA and nasopharyngeal carcinoma in Chinese—a further study. Int J Cancer. 1983;32:171–176. doi: 10.1002/ijc.2910320206. [DOI] [PubMed] [Google Scholar]
  • 13.Bei JX, Li Y, Jia WH, et al. A genome-wide association study of nasopharyngeal carcinoma identifies three new susceptibility loci. Nat Genet. 2010;42:599–603. doi: 10.1038/ng.601. [DOI] [PubMed] [Google Scholar]
  • 14.Tse KP, Su WH, Chang KP, et al. Genome-wide association study reveals multiple nasopharyngeal carcinoma-associated loci within the HLA region at chromosome 6p21.3. Am J Hum Genet. 2009;85:194–203. doi: 10.1016/j.ajhg.2009.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ng CC, Yew PY, Puah SM, et al. A genome-wide association study identifies ITGA9 conferring risk of nasopharyngeal carcinoma. J Hum Genet. 2009;54:392–397. doi: 10.1038/jhg.2009.49. [DOI] [PubMed] [Google Scholar]
  • 16.Lu CC, Chen JC, Jin YT, et al. Genetic susceptibility to naso-pharyngeal carcinoma within the HLA-A locus in Taiwanese. Int J Cancer. 2003;103:745–751. doi: 10.1002/ijc.10861. [DOI] [PubMed] [Google Scholar]
  • 17.Lu CC, Chen JC, Tsai ST, et al. Nasopharyngeal carcinoma-susceptibility locus is localized to a 132 kb segment containing HLA-A using high-resolution microsatellite mapping. Int J Cancer. 2005;115:742–746. doi: 10.1002/ijc.20946. [DOI] [PubMed] [Google Scholar]
  • 18.Lu SJ, Day NE, Degos L, et al. Linkage of a nasopharyngeal carcinoma susceptibility locus to the HLA region. Nature. 1990;346:470–471. doi: 10.1038/346470a0. [DOI] [PubMed] [Google Scholar]
  • 19.Ooi EE, Ren EC, Chan SH. Association between microsatellites within the human MHC and nasopharyngeal carcinoma. Int J Cancer. 1997;74:229–232. doi: 10.1002/(sici)1097-0215(19970422)74:2<229::aid-ijc16>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
  • 20.Cho EY, Hildesheim A, Chen CJ, et al. Nasopharyngeal carcinoma and genetic polymorphisms of DNA repair enzymes XRCC1 and hOGG1. Cancer Epidemiol Biomarkers Prev. 2003;12:1100–1104. [PubMed] [Google Scholar]
  • 21.Wu SB, Hwang SJ, Chang AS, et al. Human leukocyte antigen (HLA) frequency among patients with nasopharyngeal carcinoma in Taiwan. Anticancer Res. 1989;9:1649–1653. [PubMed] [Google Scholar]
  • 22.Yu KJ, Gao X, Chen CJ, et al. Association of human leukocyte antigens with nasopharyngeal carcinoma in high-risk multiplex families in Taiwan. Hum Immunol. 2009;70:910–914. doi: 10.1016/j.humimm.2009.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Hu SP, Day NE, Li DR, et al. Further evidence for an HLA-related recessive mutation in nasopharyngeal carcinoma among the Chinese. Br J Cancer. 2005;92:967–970. doi: 10.1038/sj.bjc.6602347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bei JX, Jia WH, Zeng YX. Familial and large-scale case-control studies identify genes associated with nasopharyngeal carcinoma. Semin Cancer Biol. 2012;22:96–106. doi: 10.1016/j.semcancer.2012.01.012. [DOI] [PubMed] [Google Scholar]
  • 25.Hildesheim A, Wang CP. Genetic predisposition factors and nasopharyngeal carcinoma risk: a review of epidemiological association studies, 2000-2011: Rosetta Stone for NPC: genetics, viral infection, and other environmental factors. Semin Cancer Biol. 2012;22:107–116. doi: 10.1016/j.semcancer.2012.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Li X, Fasano R, Wang E, et al. HLA associations with naso-pharyngeal carcinoma. Curr Mol Med. 2009;9:751–765. doi: 10.2174/156652409788970698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Burt RD, Vaughan TL, McKnight B, et al. Associations between human leukocyte antigen type and nasopharyngeal carcinoma in Caucasians in the United States. Cancer Epidemiol Biomarkers Prev. 1996;5:879–887. [PubMed] [Google Scholar]
  • 28.Jing J, Louie E, Henderson BE, et al. Histocompatibility leukocyte antigen patterns in nasopharyngeal carcinoma cases from California. Natl Cancer Inst Monogr. 1977;47:153–156. [PubMed] [Google Scholar]
  • 29.Khanna R, Burrows SR, Nicholls J, et al. Identification of cytotoxic T cell epitopes within Epstein-Barr virus (EBV) oncogene latent membrane protein 1 (LMP1): evidence for HLA A2 supertype-restricted immune recognition of EBV-infected cells by LMP1-specific cytotoxic T lymphocytes. Eur J Immunol. 1998;28:451–458. doi: 10.1002/(SICI)1521-4141(199802)28:02<451::AID-IMMU451>3.0.CO;2-U. [DOI] [PubMed] [Google Scholar]
  • 30.Lee SP, Thomas WA, Murray RJ, et al. HLA A2.1-restricted cytotoxic T cells recognizing a range of Epstein-Barr virus isolates through a defined epitope in latent membrane protein LMP2. J Virol. 1993;67:7428–7435. doi: 10.1128/jvi.67.12.7428-7435.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Lee SP, Tierney RJ, Thomas WA, et al. Conserved CTL epitopes within EBV latent membrane protein 2: a potential target for CTL-based tumor therapy. J Immunol. 1997;158:3325–3334. [PubMed] [Google Scholar]
  • 32.Butsch Kovacic M, Martin M, Gao X, et al. Variation of the killer cell immunoglobulin-like receptors and HLA-C genes in nasopharyngeal carcinoma. Cancer Epidemiol Biomarkers Prev. 2005;14:2673–2677. doi: 10.1158/1055-9965.EPI-05-0229. [DOI] [PubMed] [Google Scholar]
  • 33.Eliopoulos AG, Young LS. Activation of the cJun N-terminal kinase (JNK) pathway by the Epstein-Barr virus-encoded latent membrane protein 1 (LMP1) Oncogene. 1998;16:1731–1742. doi: 10.1038/sj.onc.1201694. [DOI] [PubMed] [Google Scholar]
  • 34.Wu L, Nakano H, Wu Z. The C-terminal activating region 2 of the Epstein-Barr virus-encoded latent membrane protein 1 activates NF-kappaB through TRAF6 and TAK1. J Biol Chem. 2006;281:2162–2169. doi: 10.1074/jbc.M505903200. [DOI] [PubMed] [Google Scholar]
  • 35.Chen CC, Liu HP, Chao M, et al. NF-kappaB-mediated transcriptional upregulation of TNFAIP2 by the Epstein-Barr virus oncoprotein, LMP1, promotes cell motility in nasopharyngeal carcinoma. Oncogene. 2014;33:3648–3659. doi: 10.1038/onc.2013.345. [DOI] [PubMed] [Google Scholar]
  • 36.Lo AK, Lo KW, Ko CW, et al. Inhibition of the LKB1-AMPK pathway by the Epstein-Barr virus-encoded LMP1 promotes proliferation and transformation of human nasopharyngeal epithelial cells. J Pathol. 2013;230:336–346. doi: 10.1002/path.4201. [DOI] [PubMed] [Google Scholar]
  • 37.Horikawa T, Yang J, Kondo S, et al. Twist and epithelial-mesenchymal transition are induced by the EBV oncoprotein latent membrane protein 1 and are associated with metastatic nasopharyngeal carcinoma. Cancer Res. 2007;67:1970–1978. doi: 10.1158/0008-5472.CAN-06-3933. [DOI] [PubMed] [Google Scholar]
  • 38.Wang Z, Luo F, Li L, et al. STAT3 activation induced by Epstein-Barr virus latent membrane protein1 causes vascular endothelial growth factor expression and cellular invasiveness via JAK3 And ERK signaling. Eur J Cancer. 2010;46:2996–3006. doi: 10.1016/j.ejca.2010.07.008. [DOI] [PubMed] [Google Scholar]
  • 39.O'Neil JD, Owen TJ, Wood VH, et al. Epstein-Barr virus-encoded EBNA1 modulates the AP-1 transcription factor pathway in nasopharyngeal carcinoma cells and enhances angiogenesis in vitro. J Gen Virol. 2008;89:2833–2842. doi: 10.1099/vir.0.2008/003392-0. [DOI] [PubMed] [Google Scholar]
  • 40.Dawson CW, Port RJ, Young LS. The role of the EBV-encoded latent membrane proteins LMP1 and LMP2 in the pathogenesis of nasopharyngeal carcinoma (NPC) Semin Cancer Biol. 2012;22:144–153. doi: 10.1016/j.semcancer.2012.01.004. [DOI] [PubMed] [Google Scholar]
  • 41.Fotheringham JA, Coalson NE, Raab-Traub N. Epstein-Barr virus latent membrane protein-2A induces ITAM/Syk- and Akt-dependent epithelial migration through alphav-integrin membrane translocation. J Virol. 2012;86:10308–10320. doi: 10.1128/JVI.00853-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Morrison JA, Klingelhutz AJ, Raab-Traub N. Epstein-Barr virus latent membrane protein 2A activates beta-catenin signaling in epithelial cells. J Virol. 2003;77:12276–12284. doi: 10.1128/JVI.77.22.12276-12284.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lun SW, Cheung ST, Cheung PF, et al. CD44+ cancer stem-like cells in EBV-associated nasopharyngeal carcinoma. PLoS One. 2012;7:e52426. doi: 10.1371/journal.pone.0052426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Port RJ, Pinheiro-Maia S, Hu C, et al. Epstein-Barr virus induction of the Hedgehog signalling pathway imposes a stem cell phenotype on human epithelial cells. J Pathol. 2013;231:367–377. doi: 10.1002/path.4245. [DOI] [PubMed] [Google Scholar]
  • 45.Cheng Y, Cheung AK, Ko JM, et al. Physiological beta-catenin signaling controls self-renewal networks and generation of stem-like cells from nasopharyngeal carcinoma. BMC Cell Biol. 2013;14:44. doi: 10.1186/1471-2121-14-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yang CF, Peng LX, Huang TJ, et al. Cancer stem-like cell characteristics induced by EB virus-encoded LMP1 contribute to radioresistance in nasopharyngeal carcinoma by suppressing the p53-mediated apoptosis pathway. Cancer Lett. 2014;344:260–271. doi: 10.1016/j.canlet.2013.11.006. [DOI] [PubMed] [Google Scholar]
  • 47.Wong AM, Kong KL, Tsang JW, et al. Profiling of Epstein-Barr virus-encoded microRNAs in nasopharyngeal carcinoma reveals potential biomarkers and oncomirs. Cancer. 2012;118:698–710. doi: 10.1002/cncr.26309. [DOI] [PubMed] [Google Scholar]
  • 48.Tsai CL, Li HP, Lu YJ, et al. Activation of DNA methyltransferase 1 by EBV LMP1 Involves c-Jun NH(2)-terminal kinase signaling. Cancer Res. 2006;66:11668–11676. doi: 10.1158/0008-5472.CAN-06-2194. [DOI] [PubMed] [Google Scholar]
  • 49.Kim JK, Samaranayake M, Pradhan S. Epigenetic mechanisms in mammals. Cell Mol Life Sci. 2009;66:596–612. doi: 10.1007/s00018-008-8432-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Li B, Huang G, Zhang X, et al. Increased phosphorylation of histone H3 at serine 10 is involved in Epstein-Barr virus latent membrane protein-1-induced carcinogenesis of nasopharyngeal carcinoma. BMC Cancer. 2013;13:124. doi: 10.1186/1471-2407-13-124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ward LD, Kellis M. Interpreting noncoding genetic variation in complex traits and human disease. Nat Biotechnol. 2012;30:1095–1106. doi: 10.1038/nbt.2422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Arvey A, Tempera I, Tsai K, et al. An atlas of the Epstein-Barr virus transcriptome and epigenome reveals host-virus regulatory interactions. Cell Host Microbe. 2012;12:233–245. doi: 10.1016/j.chom.2012.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Szeto CY, Lin CH, Choi SC, et al. Integrated mRNA and microRNA transcriptome sequencing characterizes sequence variants and mRNA-microRNA regulatory network in nasopharyngeal carcinoma model systems. FEBS Open Bio. 2014;4:128–140. doi: 10.1016/j.fob.2014.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Liu P, Fang X, Feng Z, et al. Direct sequencing and characterization of a clinical isolate of Epstein-Barr virus from nasopharyngeal carcinoma tissue by using next-generation sequencing technology. J Virol. 2011;85:11291–11299. doi: 10.1128/JVI.00823-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Li H, Homer N. A survey of sequence alignment algorithms for next-generation sequencing. Brief Bioinform. 2010;11:473–483. doi: 10.1093/bib/bbq015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Meyerson M, Gabriel S, Getz G. Advances in understanding cancer genomes through second-generation sequencing. Nat Rev Genet. 2010;11:685–696. doi: 10.1038/nrg2841. [DOI] [PubMed] [Google Scholar]
  • 57.Mwenifumbo JC, Marra MA. Cancer genome-sequencing study design. Nat Rev Genet. 2013;14:321–332. doi: 10.1038/nrg3445. [DOI] [PubMed] [Google Scholar]
  • 58.Jayasurya A, Bay BH, Yap WM, et al. Lymphocytic infiltration in undifferentiated nasopharyngeal cancer. Arch Otolaryngol Head Neck Surg. 2000;126:1329–1332. doi: 10.1001/archotol.126.11.1329. [DOI] [PubMed] [Google Scholar]
  • 59.Cibulskis K, Lawrence MS, Carter SL, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol. 2013;31:213–219. doi: 10.1038/nbt.2514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Cai MB, Han HQ, Bei JX, et al. Expression of human leukocyte antigen G is associated with prognosis in nasopharyngeal carcinoma. Int J Biol Sci. 2012;8:891–900. doi: 10.7150/ijbs.4383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Zhou W, Feng X, Ren C, et al. Over-expression of BCAT1, a c-Myc target gene, induces cell proliferation, migration and invasion in nasopharyngeal carcinoma. Mol Cancer. 2013;12:53. doi: 10.1186/1476-4598-12-53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lu QL, Elia G, Lucas S, et al. Bcl-2 proto-oncogene expression in Epstein-Barr-virus-associated nasopharyngeal carcinoma. Int J Cancer. 1993;53:29–35. doi: 10.1002/ijc.2910530107. [DOI] [PubMed] [Google Scholar]
  • 63.Sheu LF, Chen A, Meng CL, et al. Analysis of bcl-2 expression in normal, inflamed, dysplastic nasopharyngeal epithelia, and nasopharyngeal carcinoma: association with p53 expression. Hum Pathol. 1997;28:556–562. doi: 10.1016/s0046-8177(97)90078-2. [DOI] [PubMed] [Google Scholar]
  • 64.Yu Y, Dong W, Li X, et al. Significance of c-Myc and Bcl-2 protein expression in nasopharyngeal carcinoma. Arch Otolaryngol Head Neck Surg. 2003;129:1322–1326. doi: 10.1001/archotol.129.12.1322. [DOI] [PubMed] [Google Scholar]
  • 65.Hui AB, Or YY, Takano H, et al. Array-based comparative genomic hybridization analysis identified cyclin D1 as a target oncogene at 11q13.3 in nasopharyngeal carcinoma. Cancer Res. 2005;65:8125–8133. doi: 10.1158/0008-5472.CAN-05-0648. [DOI] [PubMed] [Google Scholar]
  • 66.Lai JP, Tong CL, Hong C, et al. Association between high initial tissue levels of cyclin d1 and recurrence of nasopharyngeal carcinoma. Laryngoscope. 2002;112:402–408. doi: 10.1097/00005537-200202000-00036. [DOI] [PubMed] [Google Scholar]
  • 67.Xie L, Xu L, He Z, et al. Identification of differentially expressed genes in nasopharyngeal carcinoma by means of the Atlas human cancer cDNA expression array. J Cancer Res Clin Oncol. 2000;126:400–406. doi: 10.1007/pl00008488. [DOI] [PubMed] [Google Scholar]
  • 68.Chu WK, Lee KC, Chow SE, et al. Dual regulation of the DeltaNp63 transcriptional activity by DeltaNp63 in human nasopharyngeal carcinoma cell. Biochem Biophys Res Commun. 2006;342:1356–1360. doi: 10.1016/j.bbrc.2006.02.111. [DOI] [PubMed] [Google Scholar]
  • 69.Crook T, Nicholls JM, Brooks L, et al. High level expression of deltaN-p63: a mechanism for the inactivation of p53 in undifferentiated nasopharyngeal carcinoma (NPC)? Oncogene. 2000;19:3439–3444. doi: 10.1038/sj.onc.1203656. [DOI] [PubMed] [Google Scholar]
  • 70.Guo C, Pan ZG, Li DJ, et al. The expression of p63 is associated with the differential stage in nasopharyngeal carcinoma and EBV infection. J Transl Med. 2006;4:23. doi: 10.1186/1479-5876-4-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bourouba M, Benyelles-Boufennara A, Terki N, et al. Epidermal growth factor receptor (EGFR) abundance correlates with p53 and Bcl-2 accumulation and patient age in a small cohort of North African nasopharyngeal carcinoma patients. Eur Cytokine Netw. 2011;22:38–44. doi: 10.1684/ecn.2011.0270. [DOI] [PubMed] [Google Scholar]
  • 72.Hui AB, Lo KW, Teo PM, et al. Genome wide detection of oncogene amplifications in nasopharyngeal carcinoma by array based comparative genomic hybridization. Int J Oncol. 2002;20:467–473. [PubMed] [Google Scholar]
  • 73.Leong JL, Loh KS, Putti TC, et al. Epidermal growth factor receptor in undifferentiated carcinoma of the nasopharynx. Laryngoscope. 2004;114:153–157. doi: 10.1097/00005537-200401000-00029. [DOI] [PubMed] [Google Scholar]
  • 74.Ma BB, Poon TC, To KF, et al. Prognostic significance of tumor angiogenesis, Ki 67, p53 oncoprotein, epidermal growth factor receptor and HER2 receptor protein expression in undifferentiated nasopharyngeal carcinoma—a prospective study. Head Neck. 2003;25:864–872. doi: 10.1002/hed.10307. [DOI] [PubMed] [Google Scholar]
  • 75.Miller WE, Earp HS, Raab-Traub N. The Epstein-Barr virus latent membrane protein 1 induces expression of the epidermal growth factor receptor. J Virol. 1995;69:4390–4398. doi: 10.1128/jvi.69.7.4390-4398.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wu M, Liu Y, Di X, et al. EIF4E over-expresses and enhances cell proliferation and cell cycle progression in nasopharyngeal carcinoma. Med Oncol. 2013;30:400. doi: 10.1007/s12032-012-0400-z. [DOI] [PubMed] [Google Scholar]
  • 77.Guo X, Lui WO, Qian CN, et al. Identifying cancer-related genes in nasopharyngeal carcinoma cell lines using DNA and mRNA expression profiling analyses. Int J Oncol. 2002;21:1197–1204. [PubMed] [Google Scholar]
  • 78.Fan CS, Wong N, Leung SF, et al. Frequent c-myc and Int-2 overrepresentations in nasopharyngeal carcinoma. Hum Pathol. 2000;31:169–178. doi: 10.1016/s0046-8177(00)80216-6. [DOI] [PubMed] [Google Scholar]
  • 79.Yazici H, Altun M, Alatli C, et al. c-erbB-2 gene amplification in nasopharyngeal carcinoma. Cancer Invest. 2000;18:6–10. doi: 10.3109/07357900009023056. [DOI] [PubMed] [Google Scholar]
  • 80.Porter MJ, Field JK, Leung SF, et al. The detection of the c-myc and ras oncogenes in nasopharyngeal carcinoma by immunohistochemistry. Acta Otolaryngol. 1994;114:105–109. doi: 10.3109/00016489409126025. [DOI] [PubMed] [Google Scholar]
  • 81.Yung WC, Sham JS, Choy DT, et al. ras mutations are uncommon in nasopharyngeal carcinoma. Eur J Cancer B Oral Oncol. 1995;31B:399–400. doi: 10.1016/0964-1955(95)00046-1. [DOI] [PubMed] [Google Scholar]
  • 82.Cheung HW, Ling MT, Tsao SW, et al. Id-1-induced Raf/MEK pathway activation is essential for its protective role against taxol-induced apoptosis in nasopharyngeal carcinoma cells. Carcinogenesis. 2004;25:881–887. doi: 10.1093/carcin/bgh087. [DOI] [PubMed] [Google Scholar]
  • 83.Li XJ, Peng LX, Shao JY, et al. As an independent unfavorable prognostic factor, IL-8 promotes metastasis of nasopharyngeal carcinoma through induction of epithelial-mesenchymal transition and activation of AKT signaling. Carcinogenesis. 2012;33:1302–1309. doi: 10.1093/carcin/bgs181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Meng F, Li H, Shi H, et al. MACC1 down-regulation inhibits proliferation and tumourigenicity of nasopharyngeal carcinoma cells through Akt/beta-catenin signaling pathway. PLoS One. 2013;8:e60821. doi: 10.1371/journal.pone.0060821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Qian W, Hu LF, Chen F, et al. Infrequent MDM2 gene amplification and absence of gross WAF1 gene alterations in nasopharyngeal carcinoma. Eur J Cancer B Oral Oncol. 1995;31B:328–332. doi: 10.1016/0964-1955(95)00034-8. [DOI] [PubMed] [Google Scholar]
  • 86.Wu HC, Lu TY, Lee JJ, et al. MDM2 expression in EBV-infected nasopharyngeal carcinoma cells. Lab Invest. 2004;84:1547–1556. doi: 10.1038/labinvest.3700183. [DOI] [PubMed] [Google Scholar]
  • 87.Horikawa T, Sheen TS, Takeshita H, et al. Induction of c-Met proto-oncogene by Epstein-Barr virus latent membrane protein-1 and the correlation with cervical lymph node metastasis of nasopharyngeal carcinoma. Am J Pathol. 2001;159:27–33. doi: 10.1016/S0002-9440(10)61669-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Qian CN, Guo X, Cao B, et al. Met protein expression level correlates with survival in patients with late-stage nasopharyngeal carcinoma. Cancer Res. 2002;62:589–596. [PubMed] [Google Scholar]
  • 89.Wang HB, Liu GH, Zhang H, et al. Sp1 and c-Myc regulate transcription of BMI1 in nasopharyngeal carcinoma. FEBS J. 2013;280:2929–2944. doi: 10.1111/febs.12299. [DOI] [PubMed] [Google Scholar]
  • 90.Wang WJ, Wu SP, Liu JB, et al. MYC regulation of CHK1 and CHK2 promotes radioresistance in a stem cell-like population of nasopharyngeal carcinoma cells. Cancer Res. 2013;73:1219–1231. doi: 10.1158/0008-5472.CAN-12-1408. [DOI] [PubMed] [Google Scholar]
  • 91.Fang Y, Guan X, Guo Y, et al. Analysis of genetic alterations in primary nasopharyngeal carcinoma by comparative genomic hybridization. Genes Chromosomes Cancer. 2001;30:254–260. doi: 10.1002/1098-2264(2000)9999:9999<::aid-gcc1086>3.0.co;2-d. [DOI] [PubMed] [Google Scholar]
  • 92.Or YY, Hui AB, Tam KY, et al. Characterization of chromosome 3q and 12q amplicons in nasopharyngeal carcinoma cell lines. Int J Oncol. 2005;26:49–56. [PubMed] [Google Scholar]
  • 93.Yip WK, Seow HF. Activation of phosphatidylinositol 3-kinase/Akt signaling by EGF downregulates membranous E-cadherin and beta-catenin and enhances invasion in nasopharyngeal carcinoma cells. Cancer Lett. 2012;318:162–172. doi: 10.1016/j.canlet.2011.12.018. [DOI] [PubMed] [Google Scholar]
  • 94.Endo K, Shackelford J, Aga M, et al. Upregulation of special AT-rich-binding protein 1 by Epstein-Barr virus latent membrane protein 1 in human nasopharyngeal cells and nasopharyngeal cancer. J Gen Virol. 2013;94:507–513. doi: 10.1099/vir.0.046243-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Fries KL, Miller WE, Raab-Traub N. Epstein-Barr virus latent membrane protein 1 blocks p53-mediated apoptosis through the induction of the A20 gene. J Virol. 1996;70:8653–8659. doi: 10.1128/jvi.70.12.8653-8659.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Lo PH, Lung HL, Cheung AK, et al. Extracellular protease ADAMTS9 suppresses esophageal and nasopharyngeal carcinoma tumor formation by inhibiting angiogenesis. Cancer Res. 2010;70:5567–5576. doi: 10.1158/0008-5472.CAN-09-4510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Lung HL, Lo PH, Xie D, et al. Characterization of a novel epige-netically-silenced, growth-suppressive gene, ADAMTS9, and its association with lymph node metastases in nasopharyngeal carcinoma. Int J Cancer. 2008;123:401–408. doi: 10.1002/ijc.23528. [DOI] [PubMed] [Google Scholar]
  • 98.Jin H, Wang X, Ying J, et al. Epigenetic identification of ADAMTS18 as a novel 16q23.1 tumor suppressor frequently silenced in esophageal, nasopharyngeal and multiple other carcinomas. Oncogene. 2007;26:7490–7498. doi: 10.1038/sj.onc.1210559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Liu XQ, Chen HK, Zhang XS, et al. Alterations of BLU, a candidate tumor suppressor gene on chromosome 3p21.3, in human nasopharyngeal carcinoma. Int J Cancer. 2003;106:60–65. doi: 10.1002/ijc.11166. [DOI] [PubMed] [Google Scholar]
  • 100.Yau WL, Lung HL, Zabarovsky ER, et al. Functional studies of the chromosome 3p21.3 candidate tumor suppressor gene BLU/ZMYND10 in nasopharyngeal carcinoma. Int J Cancer. 2006;119:2821–2826. doi: 10.1002/ijc.22232. [DOI] [PubMed] [Google Scholar]
  • 101.Cheng Y, Ho RLKY, Chan KC, et al. Anti-angiogenic pathway associations of the 3p21.3 mapped BLU gene in nasopharyngeal carcinoma. Oncogene. 2014 doi: 10.1038/onc.2014.353. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Zhou J, Ma J, Zhang BC, et al. BRD7, a novel bromodomain gene, inhibits G1-S progression by transcriptionally regulating some important molecules involved in ras/MEK/ERK and Rb/E2F pathways. J Cell Physiol. 2004;200:89–98. doi: 10.1002/jcp.20013. [DOI] [PubMed] [Google Scholar]
  • 103.Tsao SW, Liu Y, Wang X, et al. The association of E-cadherin expression and the methylation status of the E-cadherin gene in nasopharyngeal carcinoma cells. Eur J Cancer. 2003;39:524–531. doi: 10.1016/s0959-8049(02)00494-x. [DOI] [PubMed] [Google Scholar]
  • 104.Wang Y, Li J, Cui Y, et al. CMTM3, located at the critical tumor suppressor locus 16q22.1, is silenced by CpG methylation in carcinomas and inhibits tumor cell growth through inducing apoptosis. Cancer Res. 2009;69:5194–5201. doi: 10.1158/0008-5472.CAN-08-3694. [DOI] [PubMed] [Google Scholar]
  • 105.Cheung AK, Ko JM, Lung HL, et al. Cysteine-rich intestinal protein 2 (CRIP2) acts as a repressor of NF-kappaB-mediated proangiogenic cytokine transcription to suppress tumorigenesis and angiogenesis. Proc Natl Acad Sci U S A. 2011;108:8390–8395. doi: 10.1073/pnas.1101747108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Huang Z, Cheng Y, Chiu PM, et al. Tumor suppressor Alpha B-crystallin (CRYAB) associates with the cadherin/catenin adherens junction and impairs NPC progression-associated properties. Oncogene. 2012;31:3709–3720. doi: 10.1038/onc.2011.529. [DOI] [PubMed] [Google Scholar]
  • 107.Seng TJ, Low JS, Li H, et al. The major 8p22 tumor suppressor DLC1 is frequently silenced by methylation in both endemic and sporadic nasopharyngeal, esophageal, and cervical carcinomas, and inhibits tumor cell colony formation. Oncogene. 2007;26:934–944. doi: 10.1038/sj.onc.1209839. [DOI] [PubMed] [Google Scholar]
  • 108.Kwong J, Chow LS, Wong AY, et al. Epigenetic inactivation of the deleted in lung and esophageal cancer 1 gene in nasopharyngeal carcinoma. Genes Chromosomes Cancer. 2007;46:171–180. doi: 10.1002/gcc.20398. [DOI] [PubMed] [Google Scholar]
  • 109.Wong VC, Chen H, Ko JM, et al. Tumor suppressor dual-specificity phosphatase 6 (DUSP6) impairs cell invasion and epithelial-mesenchymal transition (EMT)-associated phenotype. Int J Cancer. 2012;130:83–95. doi: 10.1002/ijc.25970. [DOI] [PubMed] [Google Scholar]
  • 110.Law EW, Cheung AK, Kashuba VI, et al. Anti-angiogenic and tumor-suppressive roles of candidate tumor-suppressor gene, Fibulin-2, in nasopharyngeal carcinoma. Oncogene. 2012;31:728–738. doi: 10.1038/onc.2011.272. [DOI] [PubMed] [Google Scholar]
  • 111.Hwang CF, Chien CY, Huang SC, et al. Fibulin-3 is associated with tumour progression and a poor prognosis in nasopharyngeal carcinomas and inhibits cell migration and invasion via suppressed AKT activity. J Pathol. 2010;222:367–379. doi: 10.1002/path.2776. [DOI] [PubMed] [Google Scholar]
  • 112.Ying J, Srivastava G, Hsieh WS, et al. The stress-responsive gene GADD45G is a functional tumor suppressor, with its response to environmental stresses frequently disrupted epigenetically in multiple tumors. Clin Cancer Res. 2005;11:6442–6449. doi: 10.1158/1078-0432.CCR-05-0267. [DOI] [PubMed] [Google Scholar]
  • 113.Kuo YS, Tang YB, Lu TY, et al. IGFBP-6 plays a role as an oncosuppressor gene in NPC pathogenesis through regulating EGR-1 expression. J Pathol. 2010;222:299–309. doi: 10.1002/path.2735. [DOI] [PubMed] [Google Scholar]
  • 114.Lee KY, Geng H, Ng KM, et al. Epigenetic disruption of interferon-gamma response through silencing the tumor suppressor interferon regulatory factor 8 in nasopharyngeal, esophageal and multiple other carcinomas. Oncogene. 2008;27:5267–5276. doi: 10.1038/onc.2008.147. [DOI] [PubMed] [Google Scholar]
  • 115.Zhou W, Feng X, Li H, et al. Inactivation of LARS2, located at the commonly deleted region 3p21.3, by both epigenetic and genetic mechanisms in nasopharyngeal carcinoma. Acta Biochim Biophys Sin (Shanghai) 2009;41:54–62. doi: 10.1093/abbs/gmn006. [DOI] [PubMed] [Google Scholar]
  • 116.Chen H, Ko JM, Wong VC, et al. LTBP-2 confers pleiotropic suppression and promotes dormancy in a growth factor permissive microenvironment in nasopharyngeal carcinoma. Cancer Lett. 2012;325:89–98. doi: 10.1016/j.canlet.2012.06.005. [DOI] [PubMed] [Google Scholar]
  • 117.Cheung AK, Lung HL, Ko JM, et al. Chromosome 14 transfer and functional studies identify a candidate tumor suppressor gene, mirror image polydactyly 1, in nasopharyngeal carcinoma. Proc Natl Acad Sci U S A. 2009;106:14478–14483. doi: 10.1073/pnas.0900198106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Chan KC, Ko JM, Lung HL, et al. Catalytic activity of Matrix metalloproteinase-19 is essential for tumor suppressor and anti-angiogenic activities in nasopharyngeal carcinoma. Int J Cancer. 2011;129:1826–1837. doi: 10.1002/ijc.25855. [DOI] [PubMed] [Google Scholar]
  • 119.Ying J, Li H, Seng TJ, et al. Functional epigenetics identifies a protocadherin PCDH10 as a candidate tumor suppressor for nasopharyngeal, esophageal and multiple other carcinomas with frequent methylation. Oncogene. 2006;25:1070–1080. doi: 10.1038/sj.onc.1209154. [DOI] [PubMed] [Google Scholar]
  • 120.Cheung AK, Lung HL, Hung SC, et al. Functional analysis of a cell cycle-associated, tumor-suppressive gene, protein tyrosine phosphatase receptor type G, in nasopharyngeal carcinoma. Cancer Res. 2008;68:8137–8145. doi: 10.1158/0008-5472.CAN-08-0904. [DOI] [PubMed] [Google Scholar]
  • 121.Lo KW, Kwong J, Hui AB, et al. High frequency of promoter hypermethylation of RASSF1A in nasopharyngeal carcinoma. Cancer Res. 2001;61:3877–3881. [PubMed] [Google Scholar]
  • 122.Lung HL, Bangarusamy DK, Xie D, et al. THY1 is a candidate tumour suppressor gene with decreased expression in metastatic nasopharyngeal carcinoma. Oncogene. 2005;24:6525–6532. doi: 10.1038/sj.onc.1208812. [DOI] [PubMed] [Google Scholar]
  • 123.Lung HL, Cheung AK, Cheng Y, et al. Functional characterization of THY1 as a tumor suppressor gene with antiinvasive activity in nasopharyngeal carcinoma. Int J Cancer. 2010;127:304–312. doi: 10.1002/ijc.25047. [DOI] [PubMed] [Google Scholar]
  • 124.Lung HL, Cheng Y, Kumaran MK, et al. Fine mapping of the 11q22-23 tumor suppressive region and involvement of TSLC1 in nasopharyngeal carcinoma. Int J Cancer. 2004;112:628–635. doi: 10.1002/ijc.20454. [DOI] [PubMed] [Google Scholar]
  • 125.Lung HL, Cheung AK, Xie D, et al. TSLC1 is a tumor suppressor gene associated with metastasis in nasopharyngeal carcinoma. Cancer Res. 2006;66:9385–9392. doi: 10.1158/0008-5472.CAN-06-0590. [DOI] [PubMed] [Google Scholar]
  • 126.Chan SL, Cui Y, van Hasselt A, et al. The tumor suppressor Wnt inhibitory factor 1 is frequently methylated in nasopharyngeal and esophageal carcinomas. Lab Invest. 2007;87:644–650. doi: 10.1038/labinvest.3700547. [DOI] [PubMed] [Google Scholar]
  • 127.Cheng Y, Geng H, Cheng SH, et al. KRAB zinc finger protein ZNF382 is a proapoptotic tumor suppressor that represses multiple oncogenes and is commonly silenced in multiple carcinomas. Cancer Res. 2010;70:6516–6526. doi: 10.1158/0008-5472.CAN-09-4566. [DOI] [PubMed] [Google Scholar]
  • 128.Feng BJ, Huang W, Shugart YY, et al. Genome-wide scan for familial nasopharyngeal carcinoma reveals evidence of linkage to chromosome 4. Nat Genet. 2002;31:395–399. doi: 10.1038/ng932. [DOI] [PubMed] [Google Scholar]
  • 129.Xiong W, Zeng ZY, Xia JH, et al. A susceptibility locus at chromosome 3p21 linked to familial nasopharyngeal carcinoma. Cancer Res. 2004;64:1972–1974. doi: 10.1158/0008-5472.can-03-3253. [DOI] [PubMed] [Google Scholar]
  • 130.Guo XC, Scott K, Liu Y, et al. Genetic factors leading to chronic Epstein-Barr virus infection and nasopharyngeal carcinoma in South East China: study design, methods and feasibility. Hum Genomics. 2006;2:365–375. doi: 10.1186/1479-7364-2-6-365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Li Y, Fu L, Wong AM, et al. Identification of genes with allelic imbalance on 6p associated with nasopharyngeal carcinoma in southern Chinese. PLoS One. 2011;6:e14562. doi: 10.1371/journal.pone.0014562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Ghandri N, Gabbouj S, Farhat K, et al. Association of HLA-G polymorphisms with nasopharyngeal carcinoma risk and clinical outcome. Hum Immunol. 2011;72:150–158. doi: 10.1016/j.humimm.2010.10.006. [DOI] [PubMed] [Google Scholar]
  • 133.Chen Y, Chan SH. Polymorphism of T-cell receptor genes in nasopharyngeal carcinoma. Int J Cancer. 1994;56:830–833. doi: 10.1002/ijc.2910560613. [DOI] [PubMed] [Google Scholar]
  • 134.He JF, Jia WH, Fan Q, et al. Genetic polymorphisms of TLR3 are associated with Nasopharyngeal carcinoma risk in Cantonese population. BMC Cancer. 2007;7:194. doi: 10.1186/1471-2407-7-194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Aldred MA, Eng C. SNP'ing at nasopharyngeal cancer susceptibility: for whom the bell TOLLs. Cancer Biol Ther. 2006;5:1292–1293. doi: 10.4161/cbt.5.10.3487. [DOI] [PubMed] [Google Scholar]
  • 136.Xu YF, Liu WL, Dong JQ, et al. Sequencing of DC-SIGN promoter indicates an association between promoter variation and risk of nasopharyngeal carcinoma in cantonese. BMC Med Genet. 2010;11:161. doi: 10.1186/1471-2350-11-161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Xiao M, Qi F, Chen X, et al. Functional polymorphism of cytotoxic T-lymphocyte antigen 4 and nasopharyngeal carcinoma susceptibility in a Chinese population. Int J Immunogenet. 2010;37:27–32. doi: 10.1111/j.1744-313X.2009.00888.x. [DOI] [PubMed] [Google Scholar]
  • 138.Jalbout M, Bouaouina N, Gargouri J, et al. Polymorphism of the stress protein HSP70-2 gene is associated with the susceptibility to the nasopharyngeal carcinoma. Cancer Lett. 2003;193:75–81. doi: 10.1016/s0304-3835(02)00697-3. [DOI] [PubMed] [Google Scholar]
  • 139.Tsai MH, Chen WC, Tsai FJ. Correlation of p21 gene codon 31 polymorphism and TNF-alpha gene polymorphism with nasopharyngeal carcinoma. J Clin Lab Anal. 2002;16:146–150. doi: 10.1002/jcla.10032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Sousa H, Breda E, Santos AM, et al. Genetic risk markers for nasopharyngeal carcinoma in Portugal: tumor necrosis factor alpha -308G >A polymorphism. DNA Cell Biol. 2011;30:99–103. doi: 10.1089/dna.2010.1086. [DOI] [PubMed] [Google Scholar]
  • 141.Shen GP, Pan QH, Hong MH, et al. Human genetic variants of homologous recombination repair genes first found to be associated with Epstein-Barr virus antibody titers in healthy Cantonese. Int J Cancer. 2011;129:1459–1466. doi: 10.1002/ijc.25759. [DOI] [PubMed] [Google Scholar]
  • 142.Hildesheim A, Anderson LM, Chen CJ, et al. CYP2E1 genetic polymorphisms and risk of nasopharyngeal carcinoma in Taiwan. J Natl Cancer Inst. 1997;89:1207–1212. doi: 10.1093/jnci/89.16.1207. [DOI] [PubMed] [Google Scholar]
  • 143.Hildesheim A, Chen CJ, Caporaso NE, et al. Cytochrome P4502E1 genetic polymorphisms and risk of nasopharyngeal carcinoma: results from a case-control study conducted in Taiwan. Cancer Epidemiol Biomarkers Prev. 1995;4:607–610. [PubMed] [Google Scholar]
  • 144.Jia WH, Pan QH, Qin HD, et al. A case-control and a family-based association study revealing an association between CYP2E1 polymorphisms and nasopharyngeal carcinoma risk in Cantonese. Carcinogenesis. 2009;30:2031–2036. doi: 10.1093/carcin/bgp239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Tiwawech D, Srivatanakul P, Karalak A, et al. Cytochrome P450 2A6 polymorphism in nasopharyngeal carcinoma. Cancer Lett. 2006;241:135–141. doi: 10.1016/j.canlet.2005.10.026. [DOI] [PubMed] [Google Scholar]
  • 146.Jiang JH, Jia WH, Chen HK, et al. Genetic polymorphisms of CYP2A13 and its relationship to nasopharyngeal carcinoma in the Cantonese population. J Transl Med. 2004;2:24. doi: 10.1186/1479-5876-2-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Dodd LE, Sengupta S, Chen IH, et al. Genes involved in DNA repair and nitrosamine metabolism and those located on chromosome 14q32 are dysregulated in nasopharyngeal carcinoma. Cancer Epidemiol Biomarkers Prev. 2006;15:2216–2225. doi: 10.1158/1055-9965.EPI-06-0455. [DOI] [PubMed] [Google Scholar]
  • 148.Guo X, Zeng Y, Deng H, et al. Genetic Polymorphisms of CYP2E1, GSTP1, NQO1 and MPO and the Risk of Nasopharyngeal Carcinoma in a Han Chinese Population of Southern China. BMC Res Notes. 2010;3:212. doi: 10.1186/1756-0500-3-212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Nazar-Stewart V, Vaughan TL, Burt RD, et al. Glutathione S-transferase M1 and susceptibility to nasopharyngeal carcinoma. Cancer Epidemiol Biomarkers Prev. 1999;8:547–551. [PubMed] [Google Scholar]
  • 150.Guo X, O'Brien SJ, Zeng Y, et al. GSTM1 and GSTT1 gene deletions and the risk for nasopharyngeal carcinoma in Han Chinese. Cancer Epidemiol Biomarkers Prev. 2008;17:1760–1763. doi: 10.1158/1055-9965.EPI-08-0149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Zhuo X, Cai L, Xiang Z, et al. GSTM1 and GSTT1 polymorphisms and nasopharyngeal cancer risk: an evidence-based meta-analysis. J Exp Clin Cancer Res. 2009;28:46. doi: 10.1186/1756-9966-28-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Cao Y, Miao XP, Huang MY, et al. Polymorphisms of XRCC1 genes and risk of nasopharyngeal carcinoma in the Cantonese population. BMC Cancer. 2006;6:167. doi: 10.1186/1471-2407-6-167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Yang ZH, Du B, Wei YS, et al. Genetic polymorphisms of the DNA repair gene and risk of nasopharyngeal carcinoma. DNA Cell Biol. 2007;26:491–496. doi: 10.1089/dna.2006.0537. [DOI] [PubMed] [Google Scholar]
  • 154.Yang ZH, Dai Q, Kong XL, et al. Association of ERCC1 polymorphisms and susceptibility to nasopharyngeal carcinoma. Mol Carcinog. 2009;48:196–201. doi: 10.1002/mc.20468. [DOI] [PubMed] [Google Scholar]
  • 155.Ma BB, Hui EP, Wong SC, et al. Multicenter phase II study of gemcitabine and oxaliplatin in advanced nasopharyngeal carcinoma—correlation with excision repair cross-comple-menting-1 polymorphisms. Ann Oncol. 2009;20:1854–1859. doi: 10.1093/annonc/mdp065. [DOI] [PubMed] [Google Scholar]
  • 156.Qin HD, Shugart YY, Bei JX, et al. Comprehensive pathway-based association study of DNA repair gene variants and the risk of nasopharyngeal carcinoma. Cancer Res. 2011;71:3000–3008. doi: 10.1158/0008-5472.CAN-10-0469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Zheng MZ, Qin HD, Yu XJ, et al. Haplotype of gene Nedd4 binding protein 2 associated with sporadic nasopharyngeal carcinoma in the Southern Chinese population. J Transl Med. 2007;5:36. doi: 10.1186/1479-5876-5-36. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Chinese Journal of Cancer are provided here courtesy of BMC

RESOURCES