Open Access

Nasopharyngeal carcinoma: A review of current updates

  • Authors:
    • Lei Wu
    • Churong Li
    • Li Pan
  • View Affiliations

  • Published online on: February 20, 2018     https://doi.org/10.3892/etm.2018.5878
  • Pages: 3687-3692
  • Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Nasopharyngeal carcinoma (NPC) is a rare malignancy worldwide, but it is endemic in a few areas including Southern China, Southeast Asia, North Africa and the Arctic. The underlying mechanisms behind this remarkable geographic distribution remain unclear. Although Epstein‑Barr virus (EBV) infection has been suggested as a necessary cause of undifferentiated NPC, EBV itself is not sufficient to cause this malignancy. Other co-factors, such as environmental risk factors, and/or genetic susceptibility, may interact with EBV to play a role in the carcinogenesis of NPC. Survival rates differ significantly between NPC patients in early stages and late stages. Due to the close associations between EBV infection and NPC risk, EBV-related biomarkers have been used for early detection and screening for NPC in a few high-incidence areas. In the present review article the latest updates are discussed.

Introduction

Nasopharyngeal carcinoma (NPC) is one of the Epstein-Barr virus (EBV)-associated malignancies and has a characterized geographical distribution (1). In southern China, it is one of the major causes of morbidity and mortality. Despite the heavy public-health burden of NPC in southern China and other endemic areas, relatively little is known about the etiology and prevention of NPC. Although certain environmental exposures, including high consumption of salt-preserved fish, tobacco smoking and lack of fresh fruit and vegetable intake, are generally well accepted as NPC risk factors. To date there has been no rigorous population-based case-control study of NPC in southern China. Evidence accumulated so far indicated a probable causal role of EBV in the pathogenesis of undifferentiated NPC (the most common histological subtype of NPC) (1,2). However, despite establishing lifelong latency in the majority of humans, only a small proportion of individuals infected with EBV develop cancer. This indicates that EBV alone is not a sufficient cause for this malignancy. Environmental exposures and/or genetic risk factors likely also play a role in the pathogenesis of this tumor (3).

Despite the unknown etiology, using antibodies against EBV for early diagnosis and screening for NPC has been conducted in a few high-incidence areas in Southern China since the 1970s. Recent studies demonstrated that a combination of IgA antibodies against the Epstein-Barr nuclear antigen 1 (EBNA1/IgA) and VCA/IgA measured by enzyme-linked immunosorbent assay (ELISA) has higher sensitivity, specificity, and positive predictive value compared with the traditional method. Individuals identified as being at high risk of NPC based on EBV serological markers can be offered fiberoptic endoscopy/biopsy and close medical surveillance to enable early diagnosis of NPC and, ideally, reduced mortality. However, the cost-effectiveness of this labour-intensive strategy has yet not been proven, and new biomarkers are needed to more specifically identify the high-risk population, in order to provide screening for NPC in the general population.

Clinical features

The symptoms and signs at presentation of NPC include neck masses, epistaxis, nasal obstruction and discharge, headache, and other nonspecific indicators. Furthermore, because the cancer is located in a silent anatomic site, and NPC exhibits a higher metastatic rate (4), NPC tends to present at advanced stages (clinical stages III and IV) when diagnosed. It has been shown that >70% of patients were at advanced stage when diagnosed in clinics (5). A 10-year survival rate for NPC patients can reach 98% for stage I and 60% for stage II (6). In contrast, median survival is 3 years for patients at advanced stages (7), highlighting that improvements in diagnosis rate could help to reduce NPC mortality. Especially in high-incidence area, patients with symptoms should be clinically assessed for physical signs of the disease. The examination of the nasopharynx is firstly made by an indirect nasopharyngoscope, followed by a direct nasopharyngoscope (fiberoptic endoscope). A biopsy should be performed if a suspicious growth in the nasopharynx is detected. If the suspected tumor is not visible upon endoscopic examination then advanced imaging procedures like CT scan or MRI are preferred choices.

Risk factors

Since the malignancy was firstly reported in 1901 (8), the etiology of NPC has remained a puzzle for more than a century. Migrant studies show that when southern Chinese settle in other countries, their incidence of NPC is 10–30 times higher than that of other races, a rare pattern among malignancies suggesting a strong genetic component of NPC risk (9). A higher incidence of NPC is also observed among North African immigrants in Israel and Sweden, when compared to the native Israelis and native Swedes (10). Incidence of NPC among Chinese born in Western countries is still higher than that among Caucasians, although it is about half that of those living in China or migrating within Southeast Asia (10). In addition, compared with those born in southern France, men of French origin born in North Africa also had a higher incidence of NPC (11). The latter findings indicate that in addition to genetics, environmental factors also play an important role in NPC.

To date, established risk factors for type III NPC include Cantonese ethnicity (12), male sex (13), EBV infection, a family history of NPC, high consumption of salt-preserved fish, low intake of fresh vegetables and fruits intake, smoking, and some human leukocyte antigen (HLA) class I alleles (1417). On the other hand, other HLA genotypes and a history of infectious mononucleosis (IM) may be associated with a decreased risk (18). Further potential risk factors include high consumption of other preserved foods (19), a history of chronic respiratory tract conditions, and genetic polymorphisms in cytochrome P450 2E1 (CYP2E1), CYP2A6, glutathione S-transferase M1 (GSTM1) and GSTT1 (20,21). Less established risk factors include consumption of herbal medicine, occupational exposures to dust and formaldehyde, and nickel exposure (22).

Epstein-Barr virus

EBV, a γ-herpesvirus that infects lymphocytes and epithelial cells, establishes lifelong latency in >90% of adults globally (23). Primary infection with EBV usually occurs early in life and transmission is mainly through saliva. In high NPC incidence areas, such as Hong Kong, Taiwan and mainland China, ~60% of children have been infected by age 2, ~80% by age 6, and almost 100% by age 10 (24). In contrast, age at primary infection is relatively late in children from Western countries, such as US, Denmark and Sweden (24). Although primary infection with EBV is usually asymptomatic, it is associated with certain diseases, including IM and Burkitt's lymphoma (BL), ~30% of HL, certain subtypes of non-Hodgkin lymphoma, type III NPC, and a subset (~10%) of gastric carcinoma (25). It is estimated that ~143,000 deaths worldwide in 2010 could be attributed to EBV-associated malignancies (26).

The link between EBV and NPC was first proposed in 1966 when NPC patients were reported to have higher antibody response against an antigen that was later demonstrated as a product of EBV (27). Since then, extensive evidence suggests that EBV is a potential cause of NPC, especially type III. First, monoclonal EBV genome and viral gene products are detected in virtually all tumors in NPC-endemic areas (28), indicating that the tumors result from clonal proliferations of a single cell that is initially infected with EBV. Second, elevated IgA antibodies against EBV antigens are highly specific markers for subsequent NPC in high-incidence areas (29), while elevated EBV-neutralizing antibodies blocking B-cell infection and anti-gp350 antibodies are inversely associated with NPC risk (29). Third, the expression of viral proteins, such as latent membrane protein 1 (LMP1), LMP2, EBNA1 and EBNA2, has been demonstrated to drive tumor progression in invasive epithelial cancers. Nevertheless, EBV has never been detected in non-cancerous epithelial cells of the nasopharynx (30), and epithelial infection is much less efficient in vitro than B-lymphocyte infection (31). The viral target, complement receptor type 2 (CR2), which is presented on B cells and attaches to EBV envelop, gp 350/220, is expressed at low levels on epithelial cells (32). Therefore, other mechanisms of viral entry into epithelial cells have been postulated (33), including attachment to two additional glycoproteins, gHgL and gB (34), cell-to-cell contact, or IgA/secretory component (SC) protein mediation (35).

Early-life risk factors

Factors that could potentially alter the oncogenicity of EBV include the age and immune response at the time of primary EBV infection (36). For example, when primary infection is delayed until adolescence, the EBV-related immune response is robust and can lead to symptoms of IM, which is linked to risk of EBV-related HL in adulthood (37). In NPC-endemic areas, IM and HL are not prevalent, leading us to hypothesize that timing at infection may play a role in the development of NPC. Accumulated evidence shows that childhood exposures to certain environmental factors confer a higher risk of NPC (38). Nevertheless, the associations of factors influencing the timing of common childhood infections with the risk of NPC have not been studied. There are a few studies showing that a history of IM is associated with a lower risk of NPC, although commonly based on small number of cases. In high-risk populations, perhaps due to the fact that late infection is rare, it is difficult to estimate the association between a history of IM and NPC risk. In NPC non-endemic areas, however, the rarity of NPC makes the evaluation of this hypothesis a big challenge.

The household environment during childhood, when primary EBV infection is most probable, including number of siblings and population density of the household, could be important predictors for the immunological control of EBV and eventual EBV-related disease risk (39). Childhood family structure may serve as an indirect indicator of early infection with common childhood pathogens. For example, birth order has been linked to risk of other EBV-related malignancies, such as HL (40), and also to hepatocellular carcinoma (41). Hence, we hypothesized that very early exposure to EBV and other carcinogens may play a role in NPC pathogenesis. Studies on early childhood family structure may lend insights into whether timing of primary infection with EBV is associated with a subsequent risk of NPC.

Oral hygiene

Poor oral health, as a modifiable risk factor that is common among the elderly (42,43) has been linked to cancers of the pancreas, esophagus, stomach, and head and neck (44). In the case of NPC, periodontitis may increase inflammation and thus may increase the risk for NPC, given that inflammatory response could be one pathway of carcinogenesis promotion (45). In addition, bacterial load increases with a greater number of teeth lost, and some of the bacteria have been implicated in the production of nitrosamines, which are known carcinogens for NPC development (46). Poor oral health could also increase the risk of NPC by stimulating EBV replication, as indicated by higher viral load among individuals with periodontal disease than those without (47).

Few epidemiological studies have tried to address this research question of whether poor oral hygiene is related to NPC risk. One hospital-based case-control study in Turkey showed that infrequent tooth brushing and an increasing number of decayed teeth were associated with a higher NPC risk (48). Compared with those who brushed teeth daily, those who teeth brushed rarely had an odds ratio (OR) of 6.17 (95% CI, 3.60–10.55). However, when examining poor oral health as a risk factor for cancer in general, any positive associations could be due to residual confounding by smoking, low socioeconomic status, diet, and/or medical history. Detailed risk factor information in a large, population-based case-control study could help to facilitate the rigorous evaluation of oral health as a risk factor for NPC.

Familial aggregation

Familial clustering has been consistently reported in NPC high-incidence (49), intermediate-incidence (50), and low-incidence (51) areas. In Southern China, where NPC is endemic, >5% of incident cases reported a positive family history of NPC among the first-degree relatives (52,53). Previous case-control studies in different populations showed that ORs ranged from 2 to 20 in individuals who reported a first-degree family history of NPC compared with those with no such history (54,55). This magnitude of association is among the highest of any malignancy, suggesting that environmental factors themselves cannot fully explain the observed association. Genes and environmental exposures likely play a combined role in the etiology of NPC. An inheritance pattern that cannot be explained by activation of a single major susceptibility gene is supported by results from a complex segregation analysis of familial NPC showing that the etiology of NPC involves interaction of multiple genetic and environmental factors (56).

Whether familial NPC cases differ substantially from sporadic cases in terms of clinical features (i.e. histology, stage and prognosis), ethnicity, sex, age at diagnosis, environment risk factors, EBV serology, and/or genetic risk factors is still controversial (57). A study in the recent past showed that familial cases did not have characteristics notably distinct from sporadic cases (58). On the other hand, others found that familial NPC cases tend to be younger, and have better survival than sporadic cases (59,60). Further, significant modification is reported of the association with family history of NPC by smoking, wood fuel use, and salt-preserved fish consumption, whereas a prospective study did not find an interaction between smoking and family history of NPC. The small number of controls with a positive first-degree family history of NPC and the low power of the statistical test of heterogeneity make it difficult to draw firm conclusions about the joint effects of family history and environmental risk factors. Pooled studies with larger numbers of subjects will enable more powerful tests of such interactions.

To date, previous epidemiologic studies of NPC have been limited in number, size, scope and rigorousness of study design. Few studies have investigated the relative-specific risk among families with affected members. Because most studies have not ascertained all first-degree relatives and are not population-based, absolute NPC risks in the general population with and without a family history in NPC-endemic geographic regions, where the great majority of NPC cases occur worldwide, are largely unknown. The lack of evidence precludes cost-effectiveness modeling of screening for NPC among families with a positive history of NPC.

Genetic susceptibility

Only one genome-wide association study (GWAS) (61) utilized samples from >1,000 cases and controls; other GWAS were limited to a few hundred cases and controls. Many investigators have focused on the possible pathogenetic role of HLA molecules, which are required for the presentation of foreign antigens, including viral peptides, to the immune system for targeted lysis. In Chinese and other high-risk Asian populations, HLA-A2-B46, and B17 are associated with a 2- to 3-fold increase in NPC risk, whereas an increased risk is associated with HLA-B5 in Caucasians. One-third to one-half lower risk is found in association with HLA-A11 across all races, B13 in Chinese and Tunisians, and A2 in non-Chinese. Several other HLA associations have been reported, but must be interpreted with caution due to multiple-testing considerations. Genetic polymorphisms other than HLA are also reported. However, most genetic association studies are based on small sample sizes, and the lack of replication precludes a full understanding of genetic influences on NPC development.

Screening in high-incidence areas

Several lines of evidence support the notion that testing for antibodies against EBV could a useful screening tool to facilitate the early detection of NPC. First, EBV infection is an early event during tumor progression (62), and the EBV genome and gene products can be detected in virtually all tumors of type III NPC. Second, VCA/IgA neutralizing antibodies against EBV DNAse), and EA/IgA could be detected in serum even years prior to clinical evidence of the cancer, making them the basis for successful NPC screening tests in high-incidence areas. A few pilot efforts have been made to conduct NPC mass screening in high-incidence counties in southern China since the 1970s (29), using the two biomarkers of VCA/IgA and EA/IgA measured by immunofluorescence assays. More recently, studies in southern China demonstrated that a combination of EBNA1/IgA and VCA/IgA measured by ELISA had a higher diagnostic accuracy [i.e. high sensitivity, specificity, and positive predictive value (PPV)] in both the general population and families with at least two affected relatives.

Although the value of using antibodies against EBV to facilitate NPC diagnosis is generally accepted, there are a few barriers to the implementation of screening for NPC by testing these antibodies in high-incidence populations. First, only a fraction of the ~2% of individuals with elevated titers of VCA/IgA in high-risk areas develop NPC. Second, serologic evidence of EBV reactivation from latency, as indicated by elevated antibody titers against viral lytic antigens, can also be detected in normal individuals, particularly during periods of psychological or physical stress (63), thereby decreasing their specificity. Third, previous efforts were not carefully controlled (i.e. no randomized controlled trial has yet been conducted) and do not permit accurate quantification of the impact of EBV-based screening on detection rates of early-stage NPC and on NPC mortality. These results are required to support evidence-based decisions regarding the efficacy and cost-effectiveness of such screening strategies. Other than biomarkers related to EBV, biomarkers related to the human proteome may also exhibit great potential for early diagnosis of NPC. A handful of studies have used proteomics to investigate potential biomarkers for early diagnosis of NPC (64,65). Although a number of biomarkers have been identified, few have been replicated in other independent studies. Most of them may have biological implications rather than diagnostic capacity. Limited sample size, inappropriate study design, heterogeneity of NPC, and different technologies used across different studies may contribute to the inconsistency of findings.

Conclusion

To date, the technology most commonly used for human protein biomarker discovery is mass spectrometry (MS), which is limited to the analysis of a relatively small number of samples in parallel. Recently, plasma antibody profiling technologies, such as antibody suspension bead array assays have been developed for multiplex screening of a large number of proteins in patient cohorts. This advance may bring hope to identify and validate biomarkers for early diagnosis of NPC.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

LW was responsible for the conception and design of the study. LW and CL collected the files and revised the manuscript for important intellectual content. CL and LP analyzed and interpreted the files, and drafted the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ma BB, Hui EP and Chan AT: Investigational drugs for nasopharyngeal carcinoma. Expert Opin Investig Drugs. 26:677–685. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Lee HM, Lo KW, Wei W, Tsao SW, Chung GT, Ibrahim MH, Dawson CW, Murray PG, Paterson IC and Yap LF: Oncogenic S1P signalling in EBV-associated nasopharyngeal carcinoma activates AKT and promotes cell migration through S1P receptor 3. J Pathol. 242:62–72. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Wang Y, Guo Z, Zhao Y, Jin Y, An L, Wu B, Liu Z, Chen X, Chen X, Zhou H, Wang H and Zhang W: Genetic polymorphisms of lncRNA-p53 regulatory network genes are associated with concurrent chemoradiotherapy toxicities and efficacy in nasopharyngeal carcinoma patients. Sci Rep. 7:83202017. View Article : Google Scholar : PubMed/NCBI

4 

Lo KW, To KF and Huang DP: Focus on nasopharyngeal carcinoma. Cancer Cell. 5:423–428. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Tang LQ, Li CF, Li J, Chen WH, Chen QY, Yuan LX, Lai XP, He Y, Xu YX and Hu DP: Establishment and validation of prognostic nomograms for endemic nasopharyngeal carcinoma. J Natl Cancer Inst. 108:djv2912016. View Article : Google Scholar : PubMed/NCBI

6 

Chua DT, Sham JS, Kwong DL and Au GK: Treatment outcome after radiotherapy alone for patients with stage I–II nasopharyngeal carcinoma. Cancer. 98:74–80. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Al-Sarraf M, LeBlanc M, Giri PG, Fu KK, Cooper J, Vuong T, Forastiere AA, Adams G, Sakr WA, Schuller DE, et al: Chemoradiotherapy versus radiotherapy in patients with advanced nasopharyngeal cancer: Phase III randomized Intergroup study 0099. J Clin Oncol. 16:1310–1317. 1998. View Article : Google Scholar : PubMed/NCBI

8 

Jackson C: Primary carcinoma of the nasopharynx. a table of cases. J Am Med Assoc. 37:371–377. 1901. View Article : Google Scholar

9 

Parkin DM and Iscovich J: Risk of cancer in migrants and their descendants in Israel: II. Carcinomas and germ-cell tumours. Int J Cancer. 70:654–660. 1997. View Article : Google Scholar : PubMed/NCBI

10 

Mousavi SM, Sundquist J and Hemminki K: Nasopharyngeal and hypopharyngeal carcinoma risk among immigrants in Sweden. Int J Cancer. 127:2888–2892. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Warnakulasuriya KA, Johnson NW, Linklater KM and Bell J: Cancer of mouth, pharynx and nasopharynx in Asian and Chinese immigrants resident in Thames regions. Oral Oncol. 35:471–475. 1999. View Article : Google Scholar : PubMed/NCBI

12 

Jeannel D, Ghnassia M, Hubert A, Sancho-Garnier H, Eschwège F, Crognier E and de-Thé G: Increased risk of nasopharyngeal carcinoma among males of French origin born in Maghreb (north Africa). Int J Cancer. 54:536–539. 1993. View Article : Google Scholar : PubMed/NCBI

13 

Yu MC and Yuan JM: Epidemiology of nasopharyngeal carcinoma. Semin Cancer Biol. 12:421–429. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Xie SH, Yu IT, Tse LA, Mang OW and Yue L: Sex difference in the incidence of nasopharyngeal carcinoma in Hong Kong 1983–2008: Suggestion of a potential protective role of oestrogen. Eur J Cancer. 49:150–155. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Liu YT, Dai JJ, Xu CH, Lu YK, Fan YY, Zhang XL, Zhang CX and Chen YM: Greater intake of fruit and vegetables is associated with lower risk of nasopharyngeal carcinoma in Chinese adults: A case-control study. Cancer Causes Control. 23:589–599. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Xue WQ, Qin HD, Ruan HL, Shugart YY and Jia WH: Quantitative association of tobacco smoking with the risk of nasopharyngeal carcinoma: A comprehensive meta-analysis of studies conducted between 1979 and 2011. Am J Epidemiol. 178:325–338. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Hildesheim A, Apple RJ, Chen CJ, Wang SS, Cheng YJ, Klitz W, Mack SJ, Chen IH, Hsu MM, Yang CS, et al: Association of HLA class I and II alleles and extended haplotypes with nasopharyngeal carcinoma in Taiwan. J Natl Cancer Inst. 94:1780–1789. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Tang M, Lautenberger JA, Gao X, Sezgin E, Hendrickson SL, Troyer JL, David VA, Guan L, McIntosh CE, Guo X, et al: The principal genetic determinants for nasopharyngeal carcinoma in China involve the HLA class I antigen recognition groove. PLoS Genet. 8:e10031032012. View Article : Google Scholar : PubMed/NCBI

19 

Melbye M, Ebbesen P, Levine PH and Bennike T: Early primary infection and high Epstein-Barr virus antibody titers in Greenland Eskimos at high risk for nasopharyngeal carcinoma. Int J Cancer. 34:619–623. 1984. View Article : Google Scholar : PubMed/NCBI

20 

Zheng YM, Tuppin P, Hubert A, Jeannel D, Pan YJ, Zeng Y and de Thé G: Environmental and dietary risk factors for nasopharyngeal carcinoma: A case-control study in Zangwu County, Guangxi, China. Br J Cancer. 69:508–514. 1994. View Article : Google Scholar : PubMed/NCBI

21 

Lin KT, Huang WY, Lin CC, Jen YM, Lin CS, Lo CH and Kao CH: Subsequent risk of nasopharyngeal carcinoma among patients with allergic rhinitis: A nationwide population-based cohort study. Head Neck. 37:413–417. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Hildesheim A and 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. 22:107–116. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Jia WH and Qin HD: Non-viral environmental risk factors for nasopharyngeal carcinoma: A systematic review. Semin Cancer Biol. 22:117–126. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Hjalgrim H, Friborg J and Melbye M: The epidemiology of EBV and its association with malignant diseaseHuman Herpesviruses: Biology, Therapy, and Immunoprophylaxis. Cambridge University Press; 2007, View Article : Google Scholar

25 

Dowd JB, Palermo T, Brite J, McDade TW and Aiello A: Seroprevalence of Epstein-Barr virus infection in U.S. children ages 6–19, 2003–2010. PLoS One. 8:e649212013. View Article : Google Scholar : PubMed/NCBI

26 

Rickinson AB: Co-infections, inflammation and oncogenesis: Future directions for EBV research. Semin Cancer Biol. 26:99–115. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Khan G and Hashim MJ: Global burden of deaths from Epstein-Barr virus attributable malignancies 1990–2010. Infect Agent Cancer. 9:382014. View Article : Google Scholar : PubMed/NCBI

28 

Old LJ, Boyse EA, Oettgen HF, Harven ED, Geering G, Williamson B and Clifford P: Precipitating antibody in human serum to an antigen present in cultured burkitt's lymphoma cells. Proc Natl Acad Sci USA. 56:pp. 1699–1704. 1966; View Article : Google Scholar : PubMed/NCBI

29 

Pathmanathan R, Prasad U, Sadler R, Flynn K and Raab-Traub N: Clonal proliferations of cells infected with Epstein-Barr virus in preinvasive lesions related to nasopharyngeal carcinoma. N Engl J Med. 333:693–698. 1995. View Article : Google Scholar : PubMed/NCBI

30 

Chien YC, Chen JY, Liu MY, Yang HI, Hsu MM, Chen CJ and Yang CS: Serologic markers of Epstein-Barr virus infection and nasopharyngeal carcinoma in Taiwanese men. N Engl J Med. 345:1877–1882. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Coghill AE, Bu W, Nguyen H, Hsu WL, Yu KJ, Lou PJ, Wang CP, Chen CJ, Hildesheim A and Cohen JI: High levels of antibody that neutralize B-cell infection of Epstein-Barr virus and that bind EBV gp350 are associated with a lower risk of nasopharyngeal carcinoma. Clin Cancer Res. 22:3451–3457. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Sam CK, Brooks LA, Niedobitek G, Young LS, Prasad U and Rickinson AB: Analysis of Epstein-Barr virus infection in nasopharyngeal biopsies from a group at high risk of nasopharyngeal carcinoma. Int J Cancer. 53:957–962. 1993. View Article : Google Scholar : PubMed/NCBI

33 

Li QX, Young LS, Niedobitek G, Dawson CW, Birkenbach M, Wang F and Rickinson AB: Epstein-Barr virus infection and replication in a human epithelial cell system. Nature. 356:347–350. 1992. View Article : Google Scholar : PubMed/NCBI

34 

Chesnokova LS and Hutt-Fletcher LM: Epstein-Barr virus infection mechanisms. Chin J Cancer. 33:545–548. 2014.PubMed/NCBI

35 

Hutt-Fletcher LM: Epstein-Barr virus entry. J Virol. 81:7825–7832. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Sixbey JW and Yao QY: Immunoglobulin A-induced shift of Epstein-Barr virus tissue tropism. Science. 255:1578–1580. 1992. View Article : Google Scholar : PubMed/NCBI

37 

Fleisher G, Henle W, Henle G, Lennette ET and Biggar RJ: Primary infection with Epstein-Barr virus in infants in the United States: Clinical and serologic observations. J Infect Dis. 139:553–558. 1979. View Article : Google Scholar : PubMed/NCBI

38 

Callan MF, Tan L, Annels N, Ogg GS, Wilson JD, O'Callaghan CA, Steven N, McMichael AJ and Rickinson AB: Direct visualization of antigen-specific CD8+ T cells during the primary immune response to Epstein-Barr virus in vivo. J Exp Med. 187:1395–1402. 1998. View Article : Google Scholar : PubMed/NCBI

39 

Liu Z, Fang F, Chang ET, Adami HO and Ye W: Sibship size, birth order and risk of nasopharyngeal carcinoma and infectious mononucleosis: A nationwide study in Sweden. Int J Epidemiol. 45:825–834. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Chang ET, Montgomery SM, Richiardi L, Ehlin A, Ekbom A and Lambe M: Number of siblings and risk of Hodgkin's lymphoma. Cancer Epidemiol Biomarkers Prev. 13:1236–1243. 2004.PubMed/NCBI

41 

Chatenoud L, Gallus S, Altieri A, Negri E, Talamini R, Franceschi S and La Vecchia C: Number of siblings and risk of hodgkin's and other lymphoid neoplasms. Cancer Epidemiol Biomarkers Prev. 14:5522005. View Article : Google Scholar : PubMed/NCBI

42 

Hsieh CC, Tzonou A, Zavitsanos X, Kaklamani E, Lan SJ and Trichopoulos D: Age at first establishment of chronic hepatitis B virus infection and hepatocellular carcinoma risk. A birth order study. Am J Epidemiol. 136:1115–1121. 1992. View Article : Google Scholar : PubMed/NCBI

43 

Griffin SO, Jones JA, Brunson D, Griffin PM and Bailey WD: Burden of oral disease among older adults and implications for public health priorities. Am J Public Health. 102:411–418. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Abnet CC, Qiao YL, Mark SD, Dong ZW, Taylor PR and Dawsey SM: Prospective study of tooth loss and incident esophageal and gastric cancers in China. Cancer Causes Control. 12:847–854. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Huang J, Roosaar A, Axéll T and Ye W: A prospective cohort study on poor oral hygiene and pancreatic cancer risk. Int J Cancer. 138:340–347. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Coussens LM and Werb Z: Inflammation and cancer. 420:860–867. 2002.PubMed/NCBI

47 

Meurman JH and Uittamo J: Oral micro-organisms in the etiology of cancer. Acta Odontol Scand. 66:321–326. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Wang-Johanning F, Li M, Esteva FJ, Hess KR, Yin B, Rycaj K, Plummer JB, Garza JG, Ambs S and Johanning GL: Human endogenous retrovirus type K antibodies and mRNA as serum biomarkers of early-stage breast cancer. Int J Cancer. 134:587–595. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Turkoz FP, Celenkoglu G, Dogu GG, Kalender ME, Coskun U, Alkis N, Ozkan M, Turk HM and Arslan UY: Risk factors of nasopharyngeal carcinoma in Turkey-an epidemiological survey of the Anatolian Society of Medical Oncology. Asian Pac J Cancer Prev. 12:3017–3021. 2011.PubMed/NCBI

50 

Ren ZF, Liu WS, Qin HD, Xu YF, Yu DD, Feng QS, Chen LZ, Shu XO, Zeng YX and Jia WH: Effect of family history of cancers and environmental factors on risk of nasopharyngeal carcinoma in Guangdong, China. Cancer Epidemiol. 34:419–424. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Yuan JM, Wang XL, Xiang YB, Gao YT, Ross RK and Yu MC: Non-dietary risk factors for nasopharyngeal carcinoma in Shanghai, China. Int J Cancer. 85:364–369. 2000. View Article : Google Scholar : PubMed/NCBI

52 

Olajos J, Füle E, Erfán J, Krenács L, Stelkovics E, Francz M, Lengyel E, Al-Farhat Y and Esik O: Familial clustering of nasopharyngeal carcinoma in a non-endemic geographical region. Report of two Hungarian cases and a review of the literature. Acta Otolaryngol. 125:1008–1013. 2005. View Article : Google Scholar : PubMed/NCBI

53 

Yu MC, Garabrant DH, Huang TB and Henderson BE: Occupational and other non-dietary risk factors for nasopharyngeal carcinoma in Guangzhou, China. Int J Cancer. 45:1033–1039. 1990. View Article : Google Scholar : PubMed/NCBI

54 

Liu Z, Ji MF, Huang QH, Fang F, Liu Q, Jia WH, Guo X, Xie SH, Chen F, Liu Y, et al: Two Epstein-Barr virus-related serologic antibody tests in nasopharyngeal carcinoma screening: Results from the initial phase of a cluster randomized controlled trial in Southern China. Am J Epidemiol. 177:242–250. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Zou J, Sun Q, Akiba S, Yuan Y, Zha Y, Tao Z, Wei L and Sugahara T: A case-control study of nasopharyngeal carcinoma in the high background radiation areas of Yangjiang, China. J Radiat Res. 41 Suppl:53–62. 2000. View Article : Google Scholar : PubMed/NCBI

56 

Zheng X, Yan L, Nilsson B, Eklund G and Drettner B: Epstein-Barr virus infection, salted fish and nasopharyngeal carcinoma. A case-control study in southern China. Acta Oncol. 33:867–872. 1994. View Article : Google Scholar : PubMed/NCBI

57 

Jia WH, Collins A, Zeng YX, Feng BJ, Yu XJ, Huang LX, Feng QS, Huang P, Yao MH and Shugart YY: Complex segregation analysis of nasopharyngeal carcinoma in Guangdong, China: Evidence for a multifactorial mode of inheritance (complex segregation analysis of NPC in China). Eur J Hum Genet. 13:248–252. 2005. View Article : Google Scholar : PubMed/NCBI

58 

Cao SM, Guo X, Li NW, Xiang YQ, Hong MH and Min HQ: Clinical analysis of 1,142 hospitalized cantonese patients with nasopharyngeal carcinoma. Ai Zheng. 25:204–208. 2006.(In Chinese). PubMed/NCBI

59 

Cao SM, Chen SH, Qian CN, Liu Q and Xia YF: Familial nasopharyngeal carcinomas possess distinguished clinical characteristics in southern China. Chin J Cancer Res. 26:543–549. 2014.PubMed/NCBI

60 

Ouyang PY, Su Z, Mao YP, Liang XX, Liu Q and Xie FY: Prognostic impact of family history in southern Chinese patients with undifferentiated nasopharyngeal carcinoma. Br J Cancer. 109:788–794. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Rietveld CA, Medland SE, Derringer J, Yang J, Esko T, Martin NW, Westra HJ, Shakhbazov K, Abdellaoui A, Agrawal A, et al: GWAS of 126,559 individuals identifies genetic variants associated with educational attainment. Science. 340:1467–1471. 2013. View Article : Google Scholar : PubMed/NCBI

62 

He YQ, Xue WQ, Shen GP, Tang LL, Zeng YX and Jia WH: Household inhalants exposure and nasopharyngeal carcinoma risk: A large-scale case-control study in Guangdong, China. BMC Cancer. 15:10222015. View Article : Google Scholar : PubMed/NCBI

63 

Zeng Y, Zhang LG, Li HY, Jan MG, Zhang Q, Wu YC, Wang YS and Su GR: Serological mass survey for early detection of nasopharyngeal carcinoma in Wuzhou City, China. Int J Cancer. 29:139–141. 1982. View Article : Google Scholar : PubMed/NCBI

64 

Stowe RP, Pierson DL and Barrett AD: Elevated stress hormone levels relate to Epstein-Barr virus reactivation in astronauts. Psychosom Med. 63:891–895. 2001. View Article : Google Scholar : PubMed/NCBI

65 

Xiao L, Xiao T, Wang ZM, Cho WC and Xiao ZQ: Biomarker discovery of nasopharyngeal carcinoma by proteomics. Expert Rev Proteomics. 11:215–225. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2018
Volume 15 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu L, Li C and Pan L: Nasopharyngeal carcinoma: A review of current updates. Exp Ther Med 15: 3687-3692, 2018
APA
Wu, L., Li, C., & Pan, L. (2018). Nasopharyngeal carcinoma: A review of current updates. Experimental and Therapeutic Medicine, 15, 3687-3692. https://doi.org/10.3892/etm.2018.5878
MLA
Wu, L., Li, C., Pan, L."Nasopharyngeal carcinoma: A review of current updates". Experimental and Therapeutic Medicine 15.4 (2018): 3687-3692.
Chicago
Wu, L., Li, C., Pan, L."Nasopharyngeal carcinoma: A review of current updates". Experimental and Therapeutic Medicine 15, no. 4 (2018): 3687-3692. https://doi.org/10.3892/etm.2018.5878