Skip to main content
The British Journal of Radiology logoLink to The British Journal of Radiology
. 2019 Sep 4;92(1102):20190068. doi: 10.1259/bjr.20190068

Does East meet West? Towards a unified vision of the management of Nasopharyngeal carcinoma

Elaine Johanna Limkin 1,2,1,2, Pierre Blanchard 1,3,1,3,
PMCID: PMC6774589  PMID: 31150279

Abstract

Nasopharyngeal cancer (NPC) is notable for its wide geographic variation, with incidences as high as 30 in 100,000 in endemic regions but < 1 in 100,000 worldwide. This review aims to identify areas where there could be differences in prognosis, management or outcomes among countries with high or low incidence of NPC. The incidence has generally declined both in endemic and non-endemic regions throughout the years, which may be attributed to the decrease in exposure to risk factors such as early exposure to salted fish and smoking. Ethnicity has an impact both on incidence and prognosis, with Southeast Asians having the highest incidence but also better survival. Concurrent chemoradiotherapy, with or without adjuvant and/or induction chemotherapy, is the standard of care for locoregionally advanced disease, as reflected in clinical practice guidelines. Despite improvements in management, a proportion of patients relapse. Salvage treatment is associated with significant morbidity due to the critical location of the nasopharynx and the toxicities of initial therapy. Clinical expertise is paramount, but is easier to attain in endemic regions and high volume centers where enrollment of patients in clinical trials is more feasible. Collaboration between low and high incidence countries and between low and high volume facilities is key to improving NPC prognosis worldwide.

Introduction

Nasopharyngeal cancer (NPC) is a malignant epithelial tumor most commonly arising from the Fossa of Rossenmueller. A rare cancer globally with an incidence of approximately one per 100,000 people,1 it is notable for its wide geographic variation, with incidences as high as 30 in 100,000 in endemic regions such as South China and Southeast Asia.2 WHO classifies NPC into three histologic subtypes: keratinizing squamous cell, nonkeratinizing (differentiated or undifferentiated (UCNT)), and basaloid carcinoma.3 Undifferentiated carcinoma comprises over 95% of NPC in high-incidence areas and is associated with the Epstein Barr Virus (EBV) and better survival, while squamous cell carcinoma is predominant in low-incidence regions and is associated with poorer outcomes, and may have an aetiology more akin to other head and neck (H&N) cancers.4,5 A new classification has been proposed which divides NPC into epithelial carcioma, sarcomatoid carcinoma, mixed sarcomatoid-epithelial carcinoma, and squamous cell carcinoma, which had a better correlation with overall survival (OS).6

The TNM staging has recently been modified in the AJCC eighth edition.7 There were two changes to the T classification. In the previous T4 criteria, ‘‘masticator space’’ and ‘‘infratemporal fossa’’ were used synonymously, but these have been replaced by a specific description of soft-tissue involvement, decreasing ambiguity. Adjacent muscle involvement (including medial and lateral pterygoids and prevertebral muscles) was down-staged to T2. In the N classification, the supraclavicular fossa was replaced by the caudal border of the cricoid cartilage as the border for the upper and lower neck nodes, more suited to standardly used axial cross-sectional imaging. Low neck involvement and 6 cm node size were merged into a single N3 designation. T4 and N3 disease were both designated stage IVA (formerly IVA and IVB).

With its unique distribution worldwide, the purpose of this review is to compare the management of NPC among endemic countries and those where the disease is considered rare, and to highlight differences, if any. This review aims at identifying areas where there could be differences in prognosis, treatment or outcomes among countries at high and low incidence of NPC.

Ethnicity and incidence

GLOBOCAN 20121 details the very distinct regions of high NPC risk. The disease has a global incidence of 87,000 (0.6% of all cancers) and an age-standardized rate (ASR) of 1.7 in males and 0.7 in females, but high ASRs of 6.4 and 2.4 in males and females, respectively, in Southeast Asia. ASRs are also high in Micronesia/Polynesia (2.6 and 1.3), East Asia (2.5 and 1.0), and North Africa (2.3 and 1.0). Less developed regions likewise have higher cumulative ASRs of 2.0 and 0.8 compared to more developed regions with 0.6 and 0.2.

Incidence has however not remained constant throughout the years. For instance, in Hong Kong, a decrease from 28.5 in 1980 to 1984 to 20.2 in 1995 to 1999 in males and from 11.2 to 7.8 in females has been observed, with both incidence and mortality constantly being lower in females.8 Worldwide, a decrease in incidence has likewise been seen in almost all high-incidence areas, some ethnic populations in the USA and some European countries from 1970 to 2007.9

Different ethnic populations in the same country may have vastly different incidence rates, such as in China, an endemic region, where ASR in males are as high as 26.8 in Zhongshan (Southern) and as low as 0.4 in Yangcheng County (Northeastern). In the USA, a non-endemic country, incidence rates vary widely among different ethnicities: highest in Chinese, then Southeast Asians and Pacific Islanders, and low in Black and White Americans. Migrant studies have shown that ethnicity is a strong predictor of NPC, suggesting a strong genetic predisposition in its aetiology, as well as probable cultural transmission of risk factors. For instance, in Israelis and Swedes, migrants of Asian and North African descent have increased incidence of NPC, which persisted in the second and third generations in the Israeli cohort.10,11

Ethnicity and risk factors

Several risk factors have been associated with the development of NPC. EBV is an identified factor especially for the undifferentiated subtype; however, infection alone does not define disease development as the virus infects over 90% of the general population. The eventual development of cancer is influenced by other host and environmental factors.12 Certain human leukocyte antigen (HLA) genes have been found to confer a genetic susceptibility to NPC, such as the HLA-A2 and HLA-B46 which are associated with increased susceptibility in geographical areas of high incidence.13,14 Hypotheses for the link between EBV and HLA are that HLA may incite the development of NPC by modulating the expression of EBV proteins, and that HLA may represent a genetic marker of predisposition to NPC. In addition, genetic polymorphisms of some metabolic enzyme genes i.e. CYP2E1 and GSTM1 and some DNA repair genes i.e. hOGG1 and XRCC1 have also been found to be associated with increased risk of NPC.15–17

A decreased exposure to certain risk factors, such as previous early exposure to salted fish which was commonly used in weaning infants,18,19 is likely partly responsible for the decrease in incidence of NPC in certain regions such as Singapore and Hong Kong.20 This is supported by the higher incidence rate of NPC in first generation Chinese migrants, with a gradual decrease among successive generations who become more integrated into non-Chinese cultures.21

It has been shown that cigarette smoking increases the risk of NPC, particularly the squamous or keratinizing subtype.22 A recent decrease in smoking, for instance from 20.9% in 2005 to 16.8% in 2014 in the United States,23 is likely contributory to the decrease in NPC cases, more importantly for the keratinizing subtype in non-endemic areas.24

Ethnicity and prognosis

Ethnicity likewise plays a role in prognosis. A study conducted in the USA investigating racial disparities in survival of NPC in 5,427 patients from the Surveillance, Epidemiology, and End Results (SEER) database showed a statistically significantly better disease-specific survival (DSS) for African-Americans (p = 0.02) and Asians (p = 0.01) relative to Caucasian patients.25 A similar study from the SEER database corroborated that Asians had the highest incidence rates and longest survival times, consistent across all histologic types.26 Yet another study conducted in North America corroborated that Asian ethnicity was associated with improved OS and DSS (p < 0.05) on multivariate analysis. In addition, testing the interaction of chemoradiotherapy and Asian versus non-Asian ethnicity yielded a hazard ratio (HR) of 3.9, suggesting that concurrent chemoradiotherapy (CCRT) conferred more benefit in non-Asian compared to Asian subjects, yet this did not translate to improved outcomes.27 A more recent publication focused on minority groups (Blacks, Hispanics, Asians/Pacific Islanders, American Indians/Alaska Natives) who may present with different stages at diagnosis, access to healthcare and other factors which may influence outcomes. The authors found that Non-Hispanic American Indians/Alaska Natives consistently had the worst and that non-Hispanic Asians/Pacific Islanders consistently had the best survival (p < 0.001).28

In non-endemic regions, human papillomavirus (HPV) may be a contributory factor to poorer prognosis. In Michigan, HPV-positive tumors were found to be associated with smoking, older age and had a mutually-exclusive positivity with EBV, with 94% of HPV-positive status being in whites. HPV-positive tumors exhibited worse outcomes than EBV-positive tumors, with decreased OS (HR 2.98, p = 0.01), progression-free survival (PFS) (HR 2.55, p = 0.02), and local control (LC) (HR 4.01, p = 0.03).29

Treatment options

CCRT has long been the standard of care in the treatment of NPC, following the impressive improvements in PFS and OS compared to radiotherapy (RT) alone in the landmark Intergroup 00–99 trial.30 However, this study has received some criticism due to the high proportion of patients with a well-differentiated histology enrolled (only 41% had the undifferentiated subtype), such that the results might not be applicable to endemic regions with more undifferentiated subtypes. CCRT has since been studied in endemic regions, with most showing significant improvements in outcomes. The primary endpoint of PFS was not significant in Hong Kong,31 while OS was significant in Singapore (p = 0.0061)32 and in endemic regions of China (p = 0.043).33 The update of the MAC-NPC meta-analysis which included 19 trials with 4,806 patients confirmed that the addition of concomitant chemotherapy to RT significantly improves survival in patients with locoregionally advanced NPC (HR for OS 0.79, p < 0.0001; absolute benefit at 5 years 6.3%).34 However, the most dramatic improvement remained to be in the Intergroup trial, arguably in part because the control group who received RT alone had considerably poorer outcomes compared to stage-equivalent patients treated in endemic areas.

Adjuvant chemotherapy (ACT), in addition to CCRT, has not consistently been shown to be beneficial in all trials; for instance, a randomized Phase III multicenter trial in China had 2-year failure-free survival rates (FFS) of 86 versus 84% in the CCRT +ACT versus CCRT group (HR 0·74, 95% CI 0·49–1·10; p = 0·13).35 However, CCRT +ACT was found to be of highest significant benefit for OS in the MAC-NPC meta-analysis (HR 0.65, p = 0.01) and in a network meta-analysis from the same database (HR 0.65). Induction or neoadjuvant chemotherapy (NACT) was not found to be of significant benefit in the abovementioned MAC-NPC meta-analysis (HR 0.96, 0.80–1.16). Nonetheless, the value of NACT has been recently shown in a randomized Phase III multicenter trial in China, where the use of cisplatin, fluorouracil, and docetaxel (TPF) was associated with a 3-year FFS of 80 versus 72% for the CCRT alone group (HR 0·68, p = 0·034).36 Therefore, high level evidence exists to support the use of ACT and NACT for locoregionally advanced disease.

Comparison of clinical practice guidelines

An online search was conducted for clinical practice guidelines (CPGs) on NPC using the keywords “guidelines” and “nasopharyngeal cancer”. A total of 11 were found spanning all continents, including the United Kingdom,37 Spain,38 the EHNS-ESMO-ESTRO guideline,39 Malaysia,40 Asia,41 four from Canada,42–45 and two from the United States.46,47 Recommendations are summarized in Table 1.

Table 1.

Comparison of Clinical Practice Guidelines

REGION CENTER/ GROUP/ ORGANIZATION SCREEN-
ING
EBV TESTING AT DIAGNOSIS MANAGEMENT EBV monitoring LOCAL SALVAGE TREATMENT
Stage I
T1 N0
Stage II
T1-2 N0-1
Stage III
T1-3 N0-2
Stage IVA
T4N1-2 TXN3
CANADA PRINCESS MARGARET HOSPITAL/
CANADA42
NO YES RT alone CCRT + ACTa CCRT + ACTa CCRT + ACTa YES Surgery (primary and/or neck)
ONTARIO CANADA44 CCRT ± ACTb CCRT ± ACTb
ALBERTA CANADA43 NO NO RT alone CCRT ± ACTa CCRT ± ACTa CCRT ± ACTa NO Surgery OR re-irradiation (option of SBRT)
SASKATCHEWAN45 NO NO RT alone CCRT ± ACT CCRT ± ACT CCRT ± ACT NO Multidisciplinary conference
EUROPE UNITED KINGDOM37 NO NO RT alone RT alone or CCRTc CCRTc CCRTc NO Surgery (primary and/or neck (first line) OR re-irradiation OR palliative chemo
SPAIN38 NO CONSIDER (non- keratizing/ undifferentiated type) RT alone RT alone or CCRTa CCRTa ± NACT or ACT CCRTa ± NACT or
ACT
NO Neck dissection for persistent nodes
Multidisciplinary conference for local relapse
EHNS-ESMO-
ESTRO39
NO NO RT alone CCRT CCRT CCRT NO Surgery (primary and/or neck (first line) OR re-irradiation OR palliative chemo
ASIA APHNCEP41 NO YES RT alone CCRTa ± NACT or ACT CCRTa ± NACT or ACT CCRTa ± NACT or ACT YES Surgery OR re-irradiation
MALAYSIA40 NO EBER if ambiguous histology RT alone CCRTd CCRTd CCRTd NO Surgery OR re-irradiation
UNITED STATES NCCN46 NO May be considered RT alone CCRT + NACT or ACTe CCRT + NACT or ACTe CCRT +
NACT or ACTe
May be considered Surgery and/or re-irradiation ± systemic therapy
NCI47 NO YES RT alone CCRT ± ACT CCRT ± ACT or Altered fractionation RT CCRT ± ACT or Altered fractionation RT YES Surgery OR re-irradiation

ACT, adjuvant chemotherapy; APHNCEP, Asia Pacific Head and Neck Cancer expert panel; CCRT, concurrent chemoradiotherapy; EBER, EBV encoded early RNAs; EHNS-ESMO-ESTRO, European Head and Neck Society-European Society of Medical Oncology-European Society for Therapeutic Radiology and Oncology; NACT, neoadjuvant chemotherapy; NCCN: National Comprehensive Cancer Network;NCI, National Cancer Institute; RT, radiotherapy; SBRT, stereotactic body radiotherapy.

a

Concurrent: Cisplatin 100 mg/m2 q3weeks, Adjuvant: Cisplatin 80 mg/m2 + 5 FU 1000 mg/m2 x three cycles. Option: Cisplatin weekly 40 mg/m2

b

Concurrent only: Cisplatin 20 mg/m2/day and 5FU 400 mg/m2 as a 96 h continuous infusion during weeks 1 and 5 Concurrent and Adjuvant: Cisplatin 100 mg/m2 q3weeks, then Cisplatin 80 mg/m2 + 5 FU 1000 mg/m2 x three cycles

c

Cisplatin 100 mg/m2 q3weeks, option of weekly 40 mg/m2 for older patients and/or those with significant comorbidities.

d

Cisplatin-based

e

Cisplatin for CCRT. Cisplatin-5FU or Carboplatin-5FU (category 2B) for ACT. Docetaxel/Cisplatin/5FU, Cisplatin/5FU, Cisplatin/Epirubicin/Paclitaxel, Docetaxel/Cisplatin (category 2B) for NACT.

The guidelines are generally in concordance with the exception of minor differences in certain points. No publicly available guideline recommends EBV screening for NPC. EBV testing at diagnosis is recommended by the Princess Margaret Hospital (PMH), the Asia Pacific Head and Neck Cancer Expert Panel (APHNCEP), the National Comprehensive Cancer Network (NCCN), the National Cancer Institute (NCI), by the Sociedad Española de Oncología Médica (SEOM) for the nonkeratinizing histologies, and by Malaysia in case of ambiguous histology.

For treatment, all guidelines are consistent with the recommendation of RT alone for stage I disease. There is some heterogeneity for Stage II disease, with some advocating RT alone, while others having CCRT as an option in the presence of adverse risk factors (significant nodal disease, parapharyngeal tumor extension, and high plasma EBV level (SEOM)), and others recommending CCRT outright.

For locally advanced disease, CCRT is a consensus. For stages II to IV, the recommendation of the use of ACT is likewise heterogenous. PMH advocates ACT, the UK, ESMO, and Malaysian guidelines only recommend CCRT, while the rest leave ACT as an option. The regimens for concurrent chemotherapy are either Cisplatin 100 mg/m2 every 3 weeks, or 40 mg/m2 weekly. NACT, however, is not systematically recommended, mainly because high level evidence has only recently been published. The NCCN guidelines advocate CCRT with either NACT or ACT (category 2A), with CCRT alone classified as category 2B. It is worth noting, however, that variations in recommendations may be the result of differences in selecting evidence and interpreting data.48

Intensity modulated radiotherapy (IMRT) is the recommended RT technique, as it has been shown to significantly decrease treatment sequelae such as xerostomia.49,50 Hsuing et al compared 3DCRT and IMRT plans for 14 patients and showed that the benefits of IMRT are greater if patients had at least one of five risk factors - vertical length of target >7 cm, minimal distance between target and brainstem <0.1 cm, maximal anteroposterior (AP) overlap of target and brainstem >0.6 cm, maximal AP overlap of target and spinal cord >1 cm, and vertical overlap of target and eyes > 0 cm.51 In addition, a single center study from China has shown that IMRT improves clinical outcomes, including OS.52

Clinical outcomes in various countries and continents

A Chinese study of 720 exclusively stage I patients treated with RT alone from 1990 to 2012 showed an increase in the 5-year OS from 86.7% from the period of 1990 to 1996 to 99.3% from 2008 to 2012 (p < 0.001), reflecting the effectivity of IMRT as treatment techniques evolved from 2D, then 3D, then IMRT over the years.53 In India, 143 cases of NPC treated between 2005 to 2011 had a 2-year PFS and OS of 67.2 and 79.5%, respectively, with a median follow-up of 20 months.54 Another study from the same country with 206 patients from 1994 to 2004 showed high rates of 3-year DFS, OS and LC of 64%, 82.3%, and 71.1%, respectively.55 In France, 5-year OS and PFS were 78.3 and 72.5%, and 82.7 and 68.2%, respectively, for patients receiving induction plus CCRT versus CCRT alone, with no significant differences between the two groups.56 A multicenter Italian study reported OS and DFS at 5 years of 91 and 69%, respectively,57 comparable to outcomes in endemic regions. In Canada, an epidemiologic study of patients from 1993 to 2010 showed an unchanging incidence of NPC. An average annual percent increase in the 5-year OS of 1.14% was seen, with a median 5-year OS of 62.6%.58 Conditional survival (CS), calculated after a given duration of survival using data for only individuals who have survived to that predefined time of interest and thus may be better in assessing dynamic changes in prognosis, was calculated from the SEER registry of the US from 1973 to 2007. It was shown that adjusted 5-year OS improved significantly from 36.0% in 1973–1979, 41.7% in 1980–1989, 46.6% in 1990–1999, to 54.7% in 2000–2007 (p < 0.01). Interestingly, a steady improvement in CS was seen further from the time of diagnosis, which reached a plateau only at the ninth conditional year, with an OS rate of nearly 82% for long-term NPC survivors.59

Although not easily comparable across publications, these survival outcomes seem to be similar to published studies, with 5-year OS ranging from 58 to 92% with CCRT.60 The improvements in survival over the years likely reflect progress in treatment techniques and management such as the use of advanced imaging, which improves identification of local, regional, and metastatic disease extension. However, despite these general trends, notable epidemiologic differences exist. For instance, in non-Caucasians in the US, Asians and Pacific Islanders consistently had the best survival compared to other ethnicities (p < 0.001).28 Another study found that Chinese ethnicity, versus Caucasian, is an independent and favorable prognostic factor for survival in NPC with the keratinizing histology.61 This shows that the improved survival of Chinese patients cannot simply be explained by the increased proportion of them having the undifferentiated histology, which in turn may be due to genetic differences. In fact, a study of genetic polymorphisms showed that ERCC1 C8092A was an independent predictor of PFS in Chinese NPC patients treated with cisplatin-based chemotherapy, of 7.9 versus 9.3 months (p = 0.047) for the C/C phenotype versus others. Thus, further genetic and molecular studies are awaited to further elucidate differences in outcomes among different and even within the same ethnicities.62 Table 2 summarizes the key differences of NPC between the East and the West.

Table 2.

Summary of key differences between East and West

EAST - Endemic WEST - Non-endemic
Incidence High1 Low1
Histology Mostly undifferentiated2 Higher percentage of squamous keratinizing2
Ethnicity Only endemic populations9 Both endemic migrant and autochthonous populations9–11
Preventable risk factors EBV, salted fish12,18 EBV, HPV, Smoking and alcohol12,22,29
Genetic risk factors Human leukocyte antigens (i.e. HLA-A2, HLA-B46)13,14 Depends on population
Radiosensitivity More radiosensitive4,5 Less radiosensitive4,5
Outcomes Better survival9,61 Worse survival9,61

EBV: Epstein Barr Virus; HPV: Human Papilloma Virus.

Salvage treatment for locally recurrent disease

Despite improvements in patient outcomes with the improvement in radiation and chemotherapy techniques, approximately 10% of patients still experience relapse.63 Options for salvage treatment for local recurrence (LR) include nasopharyngectomy, external beam radiotherapy, stereotactic radiosurgery, and brachytherapy, alone or in combination. Choice of treatment typically depends on the extent of recurrence, as well as availability of equipment and experience or expertise of clinicians in the aforementioned salvage therapies. Disease that relapses in less than a year is generally considered radioresistant, and should be considered for surgical resection if feasible. Recently published studies on salvage options are summarized in Table 3.

Table 3.

Recent publications on salvage treatment for locally recurrent nasopharyngeal carcinoma

Author, region/country, year N Salvage treatment Secondary treatment LC OS DFS Post- treatment complications
Wong et al,
Malaysia, 201764
15 ENPG pre-op brachytherapy 2-year 66.7 % 2-year 40% No serious intraoperative complications
Liu et al,
Fudan, China, 201765
91 ENPG radio ± chemotherapy 5-year 38.3% 5-year 30.2% No serious complication
Ng et al,
Singapore, 201666
20 open NPG 5-year LC 70% 5-year 48.9% 5-year 66.7% No major intraoperative complications
You et al,
China, 201567
144 case matched ENPG vs IMRT chemotherapy (9 ENPG, 30 IMRT) 5-year 77.1 vs 55.5%, p = .003 12.5 vs 65.3%,
p < .001
Tian et al,
China, 201668
60 IMRT chemotherapy 5-year LFFS 85.7% 5-year 67.2% 5-year DFFS 96.1% 65% at least one severe complication, 18 deaths
Chan, et al
Hongkong, 201769
38 IMRT chemotherapy 3-year 44.3% 3-year 47.2% 3-year 17.5% 73.7% ≥ 1 Grade three toxicity, three deaths
Kong et al,
Fudan, China
201870
184 IMRT chemotherapy 3-year LRFS 85.1% 3-year 46% 3-year DMFS 91.1% 53% grade III-IV toxicities
Dizman et al,
Turkey,201471
24 SBRT (FSRT) chemotherapy 3-year 21% 3-year 31% 3-year 17% one grade III temporal lobe necrosis, one death
Yan et al,
China, 201772
39 (A)
42 (B)
A: brachytherapy
B: IMRT
36 mos 23.1 vs 13.7% 3-year 30.7 vs 32.6% 25.6 vs 66.7% ≥ grade III toxicity

DFFS, distant failure-free survival; DMFS: distant metastasis-free survival;ENPG, endoscopic nasopharyngectomy; FSRT, fractionated stereotactic radiotherapy; IMRT, intensity modulated radiotherapy; LC, local control; LFFS, local failure-free survival; LRFS, local recurrence-free survival; N, number of patients; OS, overall survival; PFS, progression-free survival; SBRT: stereotactic body radiotherapy.

A prognostic index was recently published which dichotomized patients into low or high risk subgroups that correlated with OS after salvage IMRT, using cohorts from endemic regions China and Singapore, with Harrell’s C indices of 0.71 in the training and 0.72 and 0.69 in the validation cohorts.73 The index was composed of the variables recurrence gross tumor volume (GTV), age at recurrence, dose of re-irradiation >68 Gy, prior RT toxicity ≥Grade 3, and rT3 or rT4. The validity of this index was studied in France on 35 recurrent NPCs but the results were not statistically significant for predicting survival or toxicity, raising the question of the applicability of this nomogram for non-endemic populations.74

60 patients from Sun Yat Sen University in China underwent re-irradiation with full course IMRT (60 to 70 Gy) for rT1 or rT2 disease. 17 patients with bulky gross tumors or a short interval between the end of primary RT and recurrence received concurrent chemotherapy. Five-year OS was 67.2%, the volume of GTV > 20 cm and presence of significant complications were independent factors for poor survival. 66% died of radiation-induced injuries.68 A similar study in Hongkong of 38 patients with rT3 and rT4 disease69 reported 3-year OS, PFS, and LC of 47.2%, 17.5%, and 44.3%, respectively. Toxicities equal to or greater than Grade 3 was 73.7%, with three patients dying of massive epistaxis. Another study from China reports Grade 3–4 toxicity rates of 53%.70 Thus, even if IMRT leads to good tumor control, treatment-related complications are a real challenge.

Long-term outcomes of 20 patients rT1 or rT2 disease treated with open nasopharyngectomy through a maxillectomy approach was reported in Singapore. The 5-year LC, DFS and OS were 70%, 48.9%, and 66.7%, respectively. 50% of patients developed tumor recurrence despite of negative surgical margins.66 Endoscopic nasopharyngectomy (ENPG) may be more advantageous than open surgery due to less morbidities compared to the open approach. A trial of 91 patients in China who underwent ENPG resulted in 5-year OS and DFS of 38.3 and 30.2%, respectively. Severe treatment-related complications due to infection secondary to delayed wound healing included nasopharyngeal necrosis (12.1%), nasopharyngeal hemorrhage (9.9%), and temporal lobe necrosis (2.2%).65 A preliminary report from Malaysia with 15 patients with only rT3 or rT4 disease showed 2-year OS and DFS of 66.7 and 40%, respectively.64 However, only carefully selected patients are eligible for surgical resection. Those with extensive tumors (T3/T4) as well as post-operative patients with positive margins would likely undergo re-irradiation. A case-matched study of 144 patients in China compared ENPG with IMRT67 concluded that IMRT was associated with higher medical costs (€11,847.80 versus 2371.71, p < 0.001), higher treatment-related complications and deaths (34.7% versus 5.6%, p < 0.001), and lower 5-year OS (77.1% versus 55.5%, p = 0.003).

The increasing use of stereotactic body radiotherapy may be a better option for re-irradiation. 24 patients were treated with fractionated stereotactic radiotherapy with a median dose of 30 Gy in five fractions in Turkey, with 3-year LC, PFS and OS Of 21%, 17 and 31%, respectively.71 One patient exhibited grade III temporal lobe necrosis, and one died of grade IV mucositis with overlapping infection.

Brachytherapy is an appealing option; however, its use is limited by several factors such as operator dependence and disease which is relatively limited with no skull base or bony invasion. A recent study compared brachytherapy with IMRT and reported similar 3-year OS rates but significantly less toxicities with brachytherapy, with 25.6 versus 66.7% having at least grade 3 toxicity, and a 14% rate of complication-related death in the IMRT group.72 Yet another promising option is the use of heavy ion therapy, which allows precise delivery of radiation to the target while sparing adjacent organs, in addition to its higher linear energy transfer (LET) and relative biological effectiveness (RBE). A Phase I/II trial has been initiated in Shanghai, China to evaluate the efficacy and safety as well as clinical outcomes of carbon ions in recurrent NPC.75

Recurrences are considerably more difficult to manage than primary NPC, given the great risk of cumulative toxicities with the initial and salvage treatments. As seen in the publications above, both RT and surgical options come with non-negligible morbidities. In addition, the nasopharynx has traditionally been considered difficult to access surgically. Experience and publications in non-endemic regions are very limited due to the rarity of the disease, of paramount importance in disease recurrence where negative margins impact control and survival.76

Systemic therapy for recurrent and metastatic disease

Chemotherapy has been the mainstay first-line treatment for metastatic NPC. Single agent and platinum doublets have been studied, mostly in Phase II retrospective trials.77 A pivotal Phase III randomized-controlled trial using gemcitabine-cisplatin (GP) in comparison with 5FU-cisplatin (PF) as first-line treatment for recurrent or metastatic NPC has recently been published. With a median follow-up of 19.4 months, the median PFS was statistically longer in the GP regimen versus PF (7.0 versus 5.6 months, HR 0.55, p < 0.0001).78 A meta-analysis comparing four commonly used first line regimens including PF, GP, taxanes plus platinum (TP), and triplet combination showed that the triplet combination demonstrated best short-term efficacy with a highest overall response rate (ORR) (0.74; 95% CI, 0.62–0.87), but with a 1-year OS rate lower than that of the TP regimen (0.74; 95% CI, 0.61–0.87 versus 0.79,95% CI, 0.65–0.92), which still showed the best prognosis, making the necessity of triplet regimens uncertain.79

Immunotherapy has recently been found to be of potential benefit in these cases. A Phase II trial of 44 patients resulted to a 20.5% ORR and 1-year OS of 59% in multiply pretreated recurrent or metastatic NPC patients.80 Pembrolizumab has likewise been found to have an ORR of 25.9% in 27 patients.81 Large-scale studies are awaited to validate these results and determine the role of immunotherapy in metastatic NPC.

Radiotherapy target delineation

Recently, an update of consensus contouring guidelines was published for H&N squamous cell cancers, with the general rule of the GTV being expanded by 5 + 5 mm to create the clinical target volumes (CTVs) of high and intermediate doses, respectively.82 The rationale is that 10 mm accounts for tumoral microscopic spread, and this rule allows increased reproducibility of contours. However, it is notable that the nasopharynx has not been included in this guideline, and a separate one has been published for NPC.83 With the narrow therapeutic window necessitating a precarious balance between tumor control and avoidance of toxicity to adjacent critical organs at risk (OARs), adequate target volume coverage is of paramount importance as it has been shown to impact tumor control.84 Because of the proximity of the nasopharynx to the skull base and the skull foraminae, the risk of intracranial extension is significant, but coverage has not been easy to achieve while respecting the dose constraints of OARs, especially for extensive disease. Also, unlike in other H&N localizations, there is paucity of surgical data in terms of the depth and distance of infiltration of NPC. Therefore, contouring guidelines are generally more generous to avoid missing potential sites of tumor spread. The high risk CTV still follows the GTV + 5 mm margin. Notable is that 55% of the experts recommend the inclusion of the entire nasopharynx in the high risk volume. The intermediate risk CTV is also recommended to be a 5 mm expansion from the CTV1, however, several key structures are mandatory. Contours need to be adjusted and expanded to include the following: superiorly, the vomer and surrounding ethmoid sinus, the inferior part of the sphenoid sinus in T1 and T2 disease and the entire sinus in T3 or T4 tumors, the cavernous sinus in T3 and T4 tumors, the bilateral foramina ovale, foramina rotunda and foramina lacera in all stages; anteriorly, 5 mm of the posterior nasal cavity, 5 mm of the posterior maxillary sinus electively to ensure adequate coverage of the pterygo-maxillary fissure and pterygo-palatine fossae; laterally, 5 mm of pterygoid muscles, the entire parapharyngeal space; and posteriorly, the anterior 1/3 of the clivus if not involved and the whole clivus if with any involvement.

Biomarkers

The role of EBV as a diagnostic, prognostic and predictive factor in NPC has been extensively studied, notably in endemic regions. Several methods of EBV detection have been employed, including EBV immunoglobulin A/viral capsid antigen (IgA/VCA) and early antigen (IgA/EA), encoded small RNAs (EBER), and DNA. EBER in situ hybridization of biopsies is a useful tool in the diagnosis of NPC, especially for metastatic lymph nodes of unknown primary.85,86 Plasma EBV DNA levels and serum levels of EBV VCA/IgA were compared in a study conducted in Guangzhou, with results suggesting that EBV DNA is more sensitive and specific in diagnosis and monitoring of NPC patients. EBV DNA levels were undetectable in patients in clinical remission, whereas VCA/IgA levels remained high. Patients with higher TNM and T stages also had significantly higher EBV DNA levels.87 EBV VCA/IgA was likewise not significant as a prognostic biomarker in patients with undetectable pretreatment EBV DNA.88

In Hong Kong, a study which screened more than 20,000 individuals identified 300 patients with persistently elevated EBV DNA levels, who subsequently underwent further work-up. Of these, 34 were found to be afflicted with NPC. Compared to a matched historical cohort, cancers detected by screening were in the earlier stages, and had a significantly better PFS (HR 0.10, 95% CI 0.05–0.18).89 Screening, however, is foreseeably only applicable to endemic regions where the incidence is significant enough to justify the procedure.

A study involving the quantification of plasma EBV DNA levels showed that undetectable levels post treatment correlated with improved relapse-free survival. Furthermore, the same study demonstrated that increased levels of EBV DNA was associated with more advanced tumor stages and patients who relapse, 6 months earlier than clinical detection.90 Yet another study showed that the post treatment levels of EBV DNA correlated with survival rates. However, the addition of adjuvant chemotherapy with a cisplatin doublet did not improve clinical outcomes for those with high post-treatment levels.91 A retrospective study from China compared 36 patients who underwent ENPG plus CCRT versus 26 who had CCRT alone for recurrent NPC, and showed that EBV DNA levels decreased significantly for the ENPG +CCRT group (Z = −3.484, p < 0.001), and that pre-treatment EBV DNA levels correlated with OS (p < 0.026) on multivariate analysis.92

Conclusion

Due to the unique geographical distribution of NPC, more studies and publications come from endemic areas secondary to more feasible patient accrual. Trials from Asia usually have large study populations, and randomization is commonplace. With this unequal patient load, expertise of clinicians becomes an important question. NPC is an especially challenging disease to manage due to its complex anatomy with proximity of tumors to dose-limiting critical structures, as well as its complicated patterns of spread and its propensity for distant metastases. In fact, a recent study showed an approximately 20% decrease in the relative risk of death for patients treated at high volume facilities (HVFs), and that there was a progressive decline in risk of death with increasing number of NPC patients treated.93 The positive effect of being treated at HVFs is corroborated by literature on other head and neck cancers. Risk of death or progression was 90% greater for stage III and IV H&N cancers treated in low (LVF) versus high volume centers. More cases of radiotherapy protocol deviations were judged as unacceptable in LVFs (11% versus 5%; p = 0.04), although this alone did not account for the disparity in 5-year OS (51.0% versus 69.1%; p = 0.002).94 Another study confirmed that treatment at HVFs versus LVFs and at academic versus non-academic facilities resulted in improved survival (5-year OS 61.6% versus 55.5% and 52.3% versus 49.7%, respectively, p < 0.001). Similar findings were seen for post-operative patients, wherein surgical HVFs were independently associated with improved OS in patients undergoing resection and post-operative RT for H&N cancer, but this survival benefit persists only when patients remain in the same facility for RT (5-year OS 63.1% versus 49.3%, p < 0.0001).95

Therefore, it is prudent to recommend that patients be managed in centers with experience and adequate patient load, especially in non-endemic areas. Establishing regional or national referral centers where patients can be sent for management is a good option. In cannot be overemphasized that proper treatment is paramount for this complex disease, especially since options for tumor recurrence are limited and are associated with significant treatment-related morbidities. Collaboration between low and high incidence countries and between low and high volume facilities is key to improving NPC prognosis worldwide.

Contributor Information

Elaine Johanna Limkin, Email: elainelimkin@gmail.com.

Pierre Blanchard, Email: pierre.blanchard@gustaveroussy.fr.

References

  • 1.Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015; 136: E359–E386. doi: 10.1002/ijc.29210 [DOI] [PubMed] [Google Scholar]
  • 2.Chang ET, Adami H-O. The enigmatic epidemiology of nasopharyngeal carcinoma. Cancer Epidemiology Biomarkers & Prevention 2006; 15: 1765–77. doi: 10.1158/1055-9965.EPI-06-0353 [DOI] [PubMed] [Google Scholar]
  • 3.Stelow EB, Wenig BM. Update from the 4th edition of the World Health organization classification of head and neck tumours: nasopharynx. Head Neck Pathol 2017; 11: 16–22. doi: 10.1007/s12105-017-0787-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Reddy SP, Raslan WF, Gooneratne S, Kathuria S, Marks JE. Prognostic significance of keratinization in nasopharyngeal carcinoma. Am J Otolaryngol 1995; 16: 103–8. doi: 10.1016/0196-0709(95)90040-3 [DOI] [PubMed] [Google Scholar]
  • 5.Marks JE, Phillips JL, Menck HR. The National Cancer data base report on the relationship of race and national origin to the histology of nasopharyngeal carcinoma. Cancer 1998; 83: 582–8. doi: [DOI] [PubMed] [Google Scholar]
  • 6.Wang H-Y, Chang Y-L, To K-F, Hwang JSG, Mai H-Q, Feng Y-F, et al. A new prognostic histopathologic classification of nasopharyngeal carcinoma. Chin J Cancer 2016; 35: 41. doi: 10.1186/s40880-016-0103-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Amin MB, Edge SB, Greene FL, Byrd DR, Brookland RK, Washington MK et al. AJCC Cancer Staging Manual. New York: Springer; 2017. [Google Scholar]
  • 8.Lee AWM, Foo W, Mang O, Sze WM, Chappell R, Lau WH, et al. Changing epidemiology of nasopharyngeal carcinoma in Hong Kong over a 20-year period (1980-99): an encouraging reduction in both incidence and mortality. Int J Cancer 2003; 103: 680–5. doi: 10.1002/ijc.10894 [DOI] [PubMed] [Google Scholar]
  • 9.Tang L-L, Chen W-Q, Xue W-Q, He Y-Q, Zheng R-S, Zeng Y-X, et al. Global trends in incidence and mortality of nasopharyngeal carcinoma. Cancer Lett 2016; 374: 22–30. doi: 10.1016/j.canlet.2016.01.040 [DOI] [PubMed] [Google Scholar]
  • 10.Rottenberg Y, Levine H, Keinan-Boker L, Derazne E, Leiba A, Kark JD. Risk of nasopharyngeal carcinoma penetrates across immigrant generations: a migrant cohort study of 2.3 million Jewish Israeli adolescents. Int J Cancer 2017; 140: 1060–7. doi: 10.1002/ijc.30525 [DOI] [PubMed] [Google Scholar]
  • 11.Mousavi SM, Sundquist J, Hemminki K. Nasopharyngeal and hypopharyngeal carcinoma risk among immigrants in Sweden. Int J Cancer 2010; 127: 2888–92. doi: 10.1002/ijc.25287 [DOI] [PubMed] [Google Scholar]
  • 12.Ali AS, Al-Shraim M, Al-Hakami AM, Jones IM. Epstein- Barr virus: clinical and epidemiological Revisits and genetic basis of oncogenesis. Open Virol J 2015; 9: 7–28. doi: 10.2174/1874357901509010007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lu SJ, Day NE, Degos L, Lepage V, Wang PC, Chan SH, et al. Linkage of a nasopharyngeal carcinoma susceptibility locus to the HLA region. Nature 1990; 346: 470–1. doi: 10.1038/346470a0 [DOI] [PubMed] [Google Scholar]
  • 14.Li X, Fasano R, Wang E, Yao K-T, Marincola FM. HLA associations with nasopharyngeal carcinoma. Curr Mol Med 2009; 9: 751–65. doi: 10.2174/156652409788970698 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hildesheim A, Anderson LM, Chen CJ, Cheng YJ, Brinton LA, Daly AK, et al. CYP2E1 genetic polymorphisms and risk of nasopharyngeal carcinoma in Taiwan. J Natl Cancer Inst 1997; 89: 1207–12. doi: 10.1093/jnci/89.16.1207 [DOI] [PubMed] [Google Scholar]
  • 16.Kongruttanachok N, Sukdikul S, Setavarin S, Kerekhjanarong V, Supiyaphun P, Voravud N, et al. Cytochrome P450 2E1 polymorphism and nasopharyngeal carcinoma development in Thailand: a correlative study. BMC Cancer 2001; 1: 1–4. doi: 10.1186/1471-2407-1-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Cho E-Y, Hildesheim A, Chen C-J, Hsu M-M, Chen I-H, Mittl BF, et al. Nasopharyngeal carcinoma and genetic polymorphisms of DNA repair enzymes XRCC1 and hOGG1. Cancer Epidemiol Biomarkers Prev 2003; 12: 1100–4. [PubMed] [Google Scholar]
  • 18.Cao S-M, Simons MJ, Qian C-N. The prevalence and prevention of nasopharyngeal carcinoma in China. Chin J Cancer 2011; 30: 114–9. doi: 10.5732/cjc.010.10377 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Zheng YM, Tuppin P, Hubert A, Jeannel D, Pan YJ, Zeng Y, et al. Environmental and dietary risk factors for nasopharyngeal carcinoma: a case-control study in Zangwu County, Guangxi, China. Br J Cancer 1994; 69: 508–14. doi: 10.1038/bjc.1994.92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Luo J, Chia KS, Chia SE, Reilly M, Tan CS, Ye W. Secular trends of nasopharyngeal carcinoma incidence in Singapore, Hong Kong and Los Angeles Chinese populations, 1973-1997. Eur J Epidemiol 2007; 22: 513–21. doi: 10.1007/s10654-007-9148-8 [DOI] [PubMed] [Google Scholar]
  • 21.Yu WM, Hussain SSM. Incidence of nasopharyngeal carcinoma in Chinese immigrants, compared with Chinese in China and South East Asia: review. J Laryngol Otol 2009; 123: 1067–74. doi: 10.1017/S0022215109005623 [DOI] [PubMed] [Google Scholar]
  • 22.Xue W-Q, Qin H-D, Ruan H-L, Shugart YY, Jia W-H. 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 2013; 178: 325–38. doi: 10.1093/aje/kws479 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Jamal A, Homa DM, O'Connor E, Babb SD, Caraballo RS, Singh T, et al. Current cigarette smoking among adults - United States, 2005-2014. MMWR Morb Mortal Wkly Rep 2015; 64: 1233–40. doi: 10.15585/mmwr.mm6444a2 [DOI] [PubMed] [Google Scholar]
  • 24.Vaughan TL, Shapiro JA, Burt RD, Swanson GM, Berwick M, Lynch CF, et al. Nasopharyngeal cancer in a low-risk population: defining risk factors by histological type. Cancer Epidemiol Biomarkers Prev 1996; 5: 587–93. [PubMed] [Google Scholar]
  • 25.Patel VJ, Chen N-W, Resto VA. Racial and ethnic disparities in nasopharyngeal cancer survival in the United States. Otolaryngol Head Neck Surg 2017; 156: 122–31. doi: 10.1177/0194599816672625 [DOI] [PubMed] [Google Scholar]
  • 26.Wang Y, Zhang Y, Ma S. Racial differences in nasopharyngeal carcinoma in the United States. Cancer Epidemiol 2013; 37: 793–802. doi: 10.1016/j.canep.2013.08.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hamilton SN, Ho C, Laskin J, Zhai Y, Mak P, Wu J. Asian versus non-Asian outcomes in nasopharyngeal carcinoma: a North American population-based analysis. Am J Clin Oncol 2016; 39: 575–80. doi: 10.1097/COC.0000000000000091 [DOI] [PubMed] [Google Scholar]
  • 28.Challapalli SD, Simpson MC, Adjei Boakye E, Walker RJ, Antisdel JL, Ward GM, et al. Survival differences in nasopharyngeal carcinoma among racial and ethnic minority groups in the United States: a retrospective cohort study. Clin Otolaryngol 2019; 44: 14–20. doi: 10.1111/coa.13225 [DOI] [PubMed] [Google Scholar]
  • 29.Stenmark MH, McHugh JB, Schipper M, Walline HM, Komarck C, Feng FY, et al. Nonendemic HPV-positive nasopharyngeal carcinoma: association with poor prognosis. Int J Radiat Oncol Biol Phys 2014; 88: 580–8. doi: 10.1016/j.ijrobp.2013.11.246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Al-Sarraf M, LeBlanc M, Giri PG, Fu KK, Cooper J, Vuong T, et al. Chemoradiotherapy versus radiotherapy in patients with advanced nasopharyngeal cancer: phase III randomized intergroup study 0099. JCO 1998; 16: 1310–7. doi: 10.1200/JCO.1998.16.4.1310 [DOI] [PubMed] [Google Scholar]
  • 31.Chan ATC, Teo PML, Ngan RK, Leung TW, Lau WH, Zee B, et al. Concurrent chemotherapy-radiotherapy compared with radiotherapy alone in locoregionally advanced nasopharyngeal carcinoma: progression-free survival analysis of a phase III randomized trial. JCO 2002; 20: 2038–44. doi: 10.1200/JCO.2002.08.149 [DOI] [PubMed] [Google Scholar]
  • 32.Wee J, Tan EH, Tai BC, Wong HB, Leong SS, Tan T, et al. Randomized trial of radiotherapy versus concurrent chemoradiotherapy followed by adjuvant chemotherapy in patients with American Joint Committee on Cancer/International Union Against cancer stage III and IV nasopharyngeal cancer of the endemic variety. J Clin Oncol 2005; 23: 6730–8. doi: 10.1200/JCO.2005.16.790 [DOI] [PubMed] [Google Scholar]
  • 33.Chen Y, Sun Y, Liang S-B, Zong J-F, Li W-F, Chen M, et al. Progress report of a randomized trial comparing long-term survival and late toxicity of concurrent chemoradiotherapy with adjuvant chemotherapy versus radiotherapy alone in patients with stage III to IVb nasopharyngeal carcinoma from endemic regions of China. Cancer 2013; 119: 2230–8. doi: 10.1002/cncr.28049 [DOI] [PubMed] [Google Scholar]
  • 34.Blanchard P, Lee A, Marguet S, Leclercq J, Ng WT, Ma J, et al. Chemotherapy and radiotherapy in nasopharyngeal carcinoma: an update of the MAC-NPC meta-analysis. Lancet Oncol 2015; 16: 645–55. doi: 10.1016/S1470-2045(15)70126-9 [DOI] [PubMed] [Google Scholar]
  • 35.Chen L, Hu C-S, Chen X-Z, Hu G-Q, Cheng Z-B, Sun Y, et al. Concurrent chemoradiotherapy plus adjuvant chemotherapy versus concurrent chemoradiotherapy alone in patients with locoregionally advanced nasopharyngeal carcinoma: a phase 3 multicentre randomised controlled trial. Lancet Oncol 2012; 13: 163–71. doi: 10.1016/S1470-2045(11)70320-5 [DOI] [PubMed] [Google Scholar]
  • 36.Sun Y, Li W-F, Chen N-Y, Zhang N, Hu G-Q, Xie F-Y, et al. Induction chemotherapy plus concurrent chemoradiotherapy versus concurrent chemoradiotherapy alone in locoregionally advanced nasopharyngeal carcinoma: a phase 3, multicentre, randomised controlled trial. Lancet Oncol 2016; 17: 1509–20. doi: 10.1016/S1470-2045(16)30410-7 [DOI] [PubMed] [Google Scholar]
  • 37.Simo R, Robinson M, Lei M, Sibtain A, Hickey S. Nasopharyngeal carcinoma: United Kingdom National multidisciplinary guidelines. J Laryngol Otol 2016; 130(S2): S97–S103. doi: 10.1017/S0022215116000517 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Pastor M, Lopez Pousa A, Del Barco E, Perez Segura P, Astorga BG, Castelo B, et al. SEOM clinical guideline in nasopharynx cancer (2017. Clin Transl Oncol 2018; 20: 84–8. doi: 10.1007/s12094-017-1777-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chan ATC, Grégoire V, Lefebvre J-L, Licitra L, Hui EP, Leung SF, et al. Nasopharyngeal cancer: EHNS-ESMO-ESTRO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2012; 23 Suppl 7(Suppl 7): vii83–5. doi: 10.1093/annonc/mds266 [DOI] [PubMed] [Google Scholar]
  • 40. Clinical Practice Guidelines Management of Nasopharyngeal Carcinoma [Internet] {Malaysia Health Technology Assessment Section (MaHTAS) Medical Development Division, Ministry of Health Malaysia}; 2016. http://www.moh.gov.my [Google Scholar]
  • 41.D'cruz A, Lin T, Anand AK, Atmakusuma D, Calaguas MJ, Chitapanarux I, et al. Consensus recommendations for management of head and neck cancer in Asian countries: a review of international guidelines. Oral Oncol 2013; 49: 872–7. doi: 10.1016/j.oraloncology.2013.05.010 [DOI] [PubMed] [Google Scholar]
  • 42.Princess Margaret Cancer Center Clinical Practice Guidelines Head and Neck (Nasopharynx) [Internet]. 2015Sep Available from: https://www.uhn.ca/PrincessMargaret/Health./CPG_HeadNeck_Nasopharynx.pdf
  • 43.Nasopharyngeal Cancer Treatment Clinical Practice Guideline HN 003 2013 Dec; Available from: www.albertahealthservices.ca
  • 44.Thephamongkhol K, Browman G, Hodson I, Oliver T, Zuraw L. Members of the Head and Neck Cancer Disease Site Group. Chemotherapy with Radiotherapy for Nasopharyngeal Cancer. Toronto (ON): Cancer Care Ontario Program in Evidence-based Care Practice Guideline. 2013;Report No.:5-7 Version 2. [Google Scholar]
  • 45.Saskatchewan Cancer Agency Nasopharyngeal Cancer Guidelines [Internet] 2015 May. Available from: www.saskcancer.ca/Head%20and%20Neck%20CPGs%2005-15
  • 46.National Comprehensive Cancer Network Head and Neck Cancers. Version 2.2018 [Internet]. [cited 2019 Mar 1]. Available from: https://www.nccn.org/professionals/physician_gls/pdf/head-and-neck.pdf
  • 47. National Cancer Institute Nasopharyngeal Cancer Treatment (Adult) (PDQ®)–Health Professional Version [Internet]; 2019. https://www.cancer.gov/types/head-and-neck/hp/adult/nasopharyngeal-treatment-pdq [Google Scholar]
  • 48.Chen Y-P, Wang Y-Q, Li W-F, Chen L, Xu C, Lu T-X, et al. Critical evaluation of the quality and recommendations of clinical practice guidelines for nasopharyngeal carcinoma. J Natl Compr Canc Netw 2017; 15: 336–44. doi: 10.6004/jnccn.2017.0033 [DOI] [PubMed] [Google Scholar]
  • 49.Pow EHN, Kwong DLW, McMillan AS, Wong MCM, Sham JST, Leung LHT, et al. Xerostomia and quality of life after intensity-modulated radiotherapy vs. conventional radiotherapy for early-stage nasopharyngeal carcinoma: initial report on a randomized controlled clinical trial. Int J Radiat Oncol Biol Phys 2006; 66: 981–91. doi: 10.1016/j.ijrobp.2006.06.013 [DOI] [PubMed] [Google Scholar]
  • 50.Kam MKM, Leung S-F, Zee B, Chau RMC, Suen JJS, Mo F, et al. Prospective randomized study of intensity-modulated radiotherapy on salivary gland function in early-stage nasopharyngeal carcinoma patients. J Clin Oncol 2007; 25: 4873–9. doi: 10.1200/JCO.2007.11.5501 [DOI] [PubMed] [Google Scholar]
  • 51.Hsiung C-Y, Yorke ED, Chui C-S, Hunt MA, Ling CC, Huang E-Y, et al. Intensity-modulated radiotherapy versus conventional three-dimensional conformal radiotherapy for boost or salvage treatment of nasopharyngeal carcinoma. International Journal of Radiation Oncology*Biology*Physics 2002; 53: 638–47. doi: 10.1016/S0360-3016(02)02760-8 [DOI] [PubMed] [Google Scholar]
  • 52.Peng G, Wang T, Yang K-Y, Zhang S, Zhang T, Li Q, et al. A prospective, randomized study comparing outcomes and toxicities of intensity-modulated radiotherapy vs. conventional two-dimensional radiotherapy for the treatment of nasopharyngeal carcinoma. Radiother Oncol 2012; 104: 286–93. doi: 10.1016/j.radonc.2012.08.013 [DOI] [PubMed] [Google Scholar]
  • 53.Tang L-Q, Lu T-Y, Li Y, Guo S-Y, Zhong Q-Y, Zou M-S, et al. Patterns of failure and survival trends of 720 patients with stage I nasopharyngeal carcinoma diagnosed from 1990-2012: a large-scale retrospective cohort study. J Cancer 2018; 9: 1308–17. doi: 10.7150/jca.21009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Haleshappa RA, Thanky AH, Kuntegowdanahalli L, Kanakasetty GB, Dasappa L, Jacob L. Epidemiology and outcomes of nasopharyngeal carcinoma: experience from a regional cancer center in southern India. South Asian J Cancer [Internet] 2017; 6: 122–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Laskar SG, Gurram L, Gupta T, Budrukkar A, Murthy V, Agarwal JP. Outcomes in nasopharyngeal carcinoma: results from a nonendemic cohort. Indian J Cancer 2016; 53: 493–8. doi: 10.4103/0019-509X.204762 [DOI] [PubMed] [Google Scholar]
  • 56.Ou D, Blanchard P, El Khoury C, De Felice F, Even C, Levy A, et al. Induction chemotherapy with docetaxel, cisplatin and fluorouracil followed by concurrent chemoradiotherapy or chemoradiotherapy alone in locally advanced non-endemic nasopharyngeal carcinoma. Oral Oncol 2016; 62: 114–21. doi: 10.1016/j.oraloncology.2016.10.011 [DOI] [PubMed] [Google Scholar]
  • 57.Tonoli S, Alterio D, Caspiani O, Bacigalupo A, Bunkheila F, Cianciulli M, et al. Nasopharyngeal carcinoma in a low incidence European area. Strahlenther Onkol 2016; 192: 931–43. doi: 10.1007/s00066-016-1052-2 [DOI] [PubMed] [Google Scholar]
  • 58.Mifsud M, Eskander A, Irish J, Gullane P, Gilbert R, Brown D, et al. Evolving trends in head and neck Cancer epidemiology: Ontario, Canada 1993-2010. Head Neck 2017; 39: 1770–8. doi: 10.1002/hed.24829 [DOI] [PubMed] [Google Scholar]
  • 59.Lv J-W, Huang X-D, Chen Y-P, Zhou G-Q, Tang L-L, Mao Y-P, et al. A national study of survival trends and conditional survival in nasopharyngeal carcinoma: analysis of the national population-based surveillance epidemiology and end results registry. Cancer Res Treat 2018; 50: 324–34. doi: 10.4143/crt.2016.544 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Chua MLK, Wee JTS, Hui EP, Chan ATC. Nasopharyngeal carcinoma. The Lancet 2016; 387: 1012–24. doi: 10.1016/S0140-6736(15)00055-0 [DOI] [PubMed] [Google Scholar]
  • 61.S-H O, Zell JA, Ziogas A, Anton-Culver H. Epidemiology of nasopharyngeal carcinoma in the United States: improved survival of Chinese patients within the keratinizing squamous cell carcinoma histology. Ann Oncol . 2007; 18: 29–35. [DOI] [PubMed] [Google Scholar]
  • 62.Chen C, Wang F, Wang Z, Li C, Luo H, Liang Y, et al. Polymorphisms in ERCC1 C8092A predict progression-free survival in metastatic/recurrent nasopharyngeal carcinoma treated with cisplatin-based chemotherapy. Cancer Chemother Pharmacol 2013; 72: 315–22. doi: 10.1007/s00280-013-2196-8 [DOI] [PubMed] [Google Scholar]
  • 63.Yu KH, Leung SF, Tung SY, Zee B, Chua DTT, Sze WM, et al. Survival outcome of patients with nasopharyngeal carcinoma with first local failure: a study by the Hong Kong nasopharyngeal carcinoma Study Group. Head Neck 2005; 27: 397–405. doi: 10.1002/hed.20161 [DOI] [PubMed] [Google Scholar]
  • 64.Wong EHC, Liew YT, Abu Bakar MZ, Lim EYL, Prepageran N. A preliminary report on the role of endoscopic endonasal nasopharyngectomy in recurrent rT3 and RT4 nasopharyngeal carcinoma. Eur Arch Otorhinolaryngol 2017; 274: 275–81. doi: 10.1007/s00405-016-4248-2 [DOI] [PubMed] [Google Scholar]
  • 65.Liu J, Yu H, Sun X, Wang D, Gu Y, Liu Q, et al. Salvage endoscopic nasopharyngectomy for local recurrent or residual nasopharyngeal carcinoma: a 10-year experience. Int J Clin Oncol 2017; 22: 834–42. doi: 10.1007/s10147-017-1143-9 [DOI] [PubMed] [Google Scholar]
  • 66.Ng LS, Lim CM, Loh KS. Long-term outcomes of nasopharyngectomy using partial maxillectomy approach. Laryngoscope 2016; 126: 1103–7. doi: 10.1002/lary.25777 [DOI] [PubMed] [Google Scholar]
  • 67.You R, Zou X, Hua Y-J, Han F, Li L, Zhao C, et al. Salvage endoscopic nasopharyngectomy is superior to intensity-modulated radiation therapy for local recurrence of selected T1-T3 nasopharyngeal carcinoma – a case-matched comparison. Radiother Oncol 2015; 115: 399–406. doi: 10.1016/j.radonc.2015.04.024 [DOI] [PubMed] [Google Scholar]
  • 68.Tian Y-M, Guan Y, Xiao W-W, Zeng L, Liu S, Lu T-X, et al. Long-term survival and late complications in intensity-modulated radiotherapy of locally recurrent T1 to T2 nasopharyngeal carcinoma. Head Neck 2016; 38: 225–31. doi: 10.1002/hed.23880 [DOI] [PubMed] [Google Scholar]
  • 69.Chan OSH, Sze HCK, Lee MCH, Chan LLK, Chang ATY, Lee SWM, et al. Reirradiation with intensity-modulated radiotherapy for locally recurrent T3 to T4 nasopharyngeal carcinoma. Head Neck 2017; 39: 533–40. doi: 10.1002/hed.24645 [DOI] [PubMed] [Google Scholar]
  • 70.Kong F, Zhou J, Du C, He X, Kong L, Hu C, et al. Long-term survival and late complications of intensity-modulated radiotherapy for recurrent nasopharyngeal carcinoma. BMC Cancer 2018; 18: 1139. doi: 10.1186/s12885-018-5055-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Dizman A, Coskun-Breuneval M, Altinisik-Inan G, Olcay GK, Cetindag MF, Guney Y. Reirradiation with robotic stereotactic body radiotherapy for recurrent nasopharyngeal carcinoma. Asian Pac J Cancer Prev 2014; 15: 3561–6. doi: 10.7314/APJCP.2014.15.8.3561 [DOI] [PubMed] [Google Scholar]
  • 72.Yan H, Mo Z, Xiang Z, Rong D, Zhang Y, Chen G, et al. CT-guided 125I brachytherapy for locally recurrent nasopharyngeal carcinoma. J Cancer 2017; 8: 2104–13. doi: 10.7150/jca.19078 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Li YQ, Tian YM, Tan SH, Liu MZ, Kusumawidjaja G, Ong EHW, YQ L, EHW O, et al. Prognostic model for stratification of radioresistant nasopharynx carcinoma to curative salvage radiotherapy. J Clin Oncol 2018; 36: 891–9. doi: 10.1200/JCO.2017.75.5165 [DOI] [PubMed] [Google Scholar]
  • 74.Ruffier A, Tao Y, Nguyen F, Moya-Plana A, Even C, Berthold C et al. Local recurrence of nasopharyngeal – outcomes after reirradiation In: 7th International Congress on Innovative Approaches in Head and Neck Oncology. [Google Scholar]
  • 75.Kong L, Hu J, Guan X, Gao J, Lu R, Lu JJ. Phase I/II trial evaluating carbon ion radiotherapy for salvaging treatment of locally recurrent nasopharyngeal carcinoma. J Cancer 2016; 7: 774–83. doi: 10.7150/jca.14399 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Tsang RK, Wei WI. Salvage surgery for nasopharyngeal cancer. World J Otorhinolaryngol Head Neck Surg 2015; 1: 34–43. doi: 10.1016/j.wjorl.2015.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Lee V, Kwong D, Leung T-W, Lam K-O, Tong C-C, Lee A. Palliative systemic therapy for recurrent or metastatic nasopharyngeal carcinoma - How far have we achieved? Crit Rev Oncol Hematol 2017; 114: 13–23. doi: 10.1016/j.critrevonc.2017.03.030 [DOI] [PubMed] [Google Scholar]
  • 78.Zhang L, Huang Y, Hong S, Yang Y, Yu G, Jia J, et al. Gemcitabine plus cisplatin versus fluorouracil plus cisplatin in recurrent or metastatic nasopharyngeal carcinoma: a multicentre, randomised, open-label, phase 3 trial. The Lancet 2016; 388: 1883–92. doi: 10.1016/S0140-6736(16)31388-5 [DOI] [PubMed] [Google Scholar]
  • 79.Ma S-X, Zhou T, Huang Y, Yang Y-P, Zhan J-H, Zhang Y-X, et al. The efficacy of first-line chemotherapy in recurrent or metastatic nasopharyngeal carcinoma: a systematic review and meta-analysis. Ann Transl Med 2018; 6: 201. doi: 10.21037/atm.2018.05.14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ma BBY, Lim W-T, Goh B-C, Hui EP, Lo K-W, Pettinger A, et al. Antitumor activity of nivolumab in recurrent and metastatic nasopharyngeal carcinoma: an international, multicenter study of the Mayo Clinic phase 2 Consortium (NCI-9742. J Clin Oncol 2018; 36: 1412–8. doi: 10.1200/JCO.2017.77.0388 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Hsu C, Lee S-H, Ejadi S, Even C, Cohen RB, Le Tourneau C, et al. Safety and antitumor activity of pembrolizumab in patients with programmed Death-Ligand 1-positive nasopharyngeal carcinoma: results of the KEYNOTE-028 study. J Clin Oncol 2017; 35: 4050–6. doi: 10.1200/JCO.2017.73.3675 [DOI] [PubMed] [Google Scholar]
  • 82.Grégoire V, Evans M, Le Q-T, Bourhis J, Budach V, Chen A, et al. Delineation of the primary tumour clinical target volumes (CTV-P) in laryngeal, hypopharyngeal, oropharyngeal and oral cavity squamous cell carcinoma: AIRO, CACA, DAHANCA, EORTC, GEORCC, GORTEC, HKNPCSG, HNCIG, IAG-KHT, LPRHHT, NCIC CTG, NCRI, NRG oncology, PHNS, SBRT, SOMERA, SRO, SSHNO, TROG consensus guidelines. Radiother Oncol 2018; 126: 3–24Available from[Internet]. doi: 10.1016/j.radonc.2017.10.016 [DOI] [PubMed] [Google Scholar]
  • 83.Lee AW, Ng WT, Pan JJ, Poh SS, Ahn YC, AlHussain H, et al. International guideline for the delineation of the clinical target volumes (CTV) for nasopharyngeal carcinoma. Radiother Oncol 2018; 126: 25–36. doi: 10.1016/j.radonc.2017.10.032 [DOI] [PubMed] [Google Scholar]
  • 84.Ng WT, Lee MCH, Chang ATY, Chan OSH, Chan LLK, Cheung FY, et al. The impact of dosimetric inadequacy on treatment outcome of nasopharyngeal carcinoma with IMRT. Oral Oncol 2014; 50: 506–12. doi: 10.1016/j.oraloncology.2014.01.017 [DOI] [PubMed] [Google Scholar]
  • 85.Tsai ST, Jin YT, Mann RB, Ambinder RF. Epstein-Barr virus detection in nasopharyngeal tissues of patients with suspected nasopharyngeal carcinoma. Cancer 1998; 82: 1449–53. doi: [DOI] [PubMed] [Google Scholar]
  • 86.Nakao K, Yuge T, Mochiki M, Nibu K-ichi, Sugasawa M. Detection of Epstein-Barr virus in metastatic lymph nodes of patients with nasopharyngeal carcinoma and a primary unknown carcinoma. Arch Otolaryngol Head Neck Surg 2003; 129: 338. doi: 10.1001/archotol.129.3.338 [DOI] [PubMed] [Google Scholar]
  • 87.Shao J-Y, Li Y-H, Gao H-Y, Wu Q-L, Cui N-J, Zhang L, et al. Comparison of plasma Epstein-Barr virus (EBV) DNA levels and serum EBV immunoglobulin A/virus capsid antigen antibody titers in patients with nasopharyngeal carcinoma. Cancer 2004; 100: 1162–70. doi: 10.1002/cncr.20099 [DOI] [PubMed] [Google Scholar]
  • 88.Yao J-J, Lin L, Jin Y-N, Wang S-Y, Zhang W-J, Zhang F, et al. Prognostic value of serum Epstein-Barr virus antibodies in patients with nasopharyngeal carcinoma and undetectable pretreatment Epstein-Barr virus DNA. Cancer Sci 2017; 108: 1640–7. doi: 10.1111/cas.13296 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Chan KCA, Woo JKS, King A, Zee BCY, Lam WKJ, Chan SL, et al. Analysis of plasma Epstein-Barr virus DNA to screen for nasopharyngeal cancer. N Engl J Med 2017; 377: 513–22. doi: 10.1056/NEJMoa1701717 [DOI] [PubMed] [Google Scholar]
  • 90.Lin J-C, Wang W-Y, Chen KY, Wei Y-H, Liang W-M, Jan J-S, et al. Quantification of plasma Epstein-Barr virus DNA in patients with advanced nasopharyngeal carcinoma. N Engl J Med 2004; 350: 2461–70. doi: 10.1056/NEJMoa032260 [DOI] [PubMed] [Google Scholar]
  • 91.Chan ATC, Hui EP, Ngan RKC, Tung SY, Cheng ACK, Ng WT, et al. Analysis of plasma Epstein-Barr virus DNA in nasopharyngeal cancer after chemoradiation to identify high-risk patients for adjuvant chemotherapy: a randomized controlled trial. JCO 2018; 36: 3091–100. doi: 10.1200/JCO.2018.77.7847 [DOI] [PubMed] [Google Scholar]
  • 92.Weng J, Wei J, Si J, Qin Y, Li M, Liu F, et al. Clinical outcomes of residual or recurrent nasopharyngeal carcinoma treated with endoscopic nasopharyngectomy plus chemoradiotherapy or with chemoradiotherapy alone: a retrospective study. PeerJ 2017; 5(Suppl 1): e3912: e3912. doi: 10.7717/peerj.3912 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Yoshida EJ, Luu M, David JM, Kim S, Mita A, Scher K, et al. Facility volume and survival in nasopharyngeal carcinoma. International Journal of Radiation Oncology*Biology*Physics 2018; 100: 408–17. doi: 10.1016/j.ijrobp.2017.09.038 [DOI] [PubMed] [Google Scholar]
  • 94.Wuthrick EJ, Zhang Q, Machtay M, Rosenthal DI, Nguyen-Tan PF, Fortin A, et al. Institutional clinical trial accrual volume and survival of patients with head and neck cancer. JCO 2015; 33: 156–64. doi: 10.1200/JCO.2014.56.5218 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lee NCJ, Kelly JR, An Y, Park HS, Judson BL, Burtness BA, et al. Radiation therapy treatment facility and overall survival in the adjuvant setting for locally advanced head and neck squamous cell carcinoma. Cancer 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The British Journal of Radiology are provided here courtesy of Oxford University Press

RESOURCES