Malignant Nasopharyngeal Tumors

Back

Practice Essentials

External beam radiation therapy with or without chemotherapy is the primary mode of therapy for previously untreated nasopharyngeal carcinoma (NPC). Recurrent or persistent disease remains a challenge to clinicians. Typically, re-irradiation is advocated. In some institutions, salvage nasopharyngectomy is used for the treatment of recurrent disease.[1]

External beam radiation therapy is the primary mode of management of NPC, both at the primary site and in the neck. This is mainly because of this tumor's high degree of sensitivity to radiation, as well as the anatomical constraints for surgical access to the highly complex nasopharyngeal region.

Management of locally recurrent diseases can be accomplished with either re-irradiation or salvage nasopharyngectomy. Patients whose treatment failed regionally can be treated with either re-irradiation or salvage neck dissection. A high prevalence of distant metastases has been observed for patients with NPC.

History of the Procedure

In 1988, Fee and Tu published results of salvage nasopharyngectomy in a series of patients with recurrent NPC who failed previous treatment with radiation.[2, 3] The results were encouraging[2] and inspired other investigators to start using surgery in the treatment of patients with recurrent NPC. Since then, various surgical approaches to the nasopharynx have been proposed. These include the transpalatal-maxillary-cervical, maxillary swing, transmandibular, transcervico-mandibulo-palatal, infratemporal fossa, lateral temporal, endoscopic, and robotic approaches.

Problem

Despite recent advances in the management of NPC, locoregional failure is still significant, with reported rates of 15.6-58% (median, 34%).[4, 5, 6, 7] Salvage treatment for local failure continues to be challenging because of the proximity of tumors to vital structures.

Epidemiology

Frequency and mortality

Nasopharyngeal carcinoma (NPC) is a prevalent malignancy in Southeast Asia.[8] In areas such as southern China, Hong Kong, Singapore, Malaysia, and Taiwan, the reported incidence rate ranges from 10-53 cases per 100,000 persons per year. The incidence is also high among Eskimos in Alaska and Greenland and in Tunisians, ranging from 15-20 cases per 100,000 persons per year. Although NPC is a relatively uncommon disease in Western countries (< 1 case per 100,000 persons), it poses a significant health problem in regions of the United States with large Asian populations. The prevalence rate for people of Asian descent in the United States is 3.0-4.2 cases per 100,000 persons.

A study by Tang et al found that between 1970 and 2007, the age-standardized incidence rate of NPC saw significant reductions in southern and eastern Asia, North America, and the Nordic nations, with the average annual percent declines being 0.9-5.4% in males and 1.1-4.1% in females. Age-standardized mortality rates between 1970 and 2013 also fell, showing average annual percent reductions of 0.9-3.7% in males and 0.8-6.5% in females. The investigators suggested that the incidence reductions were associated with tobacco control, dietary changes, and economic development, while the drop in mortality rates stemmed not only from the reduced incidence but also from diagnostic advancements and improved radiotherapy techniques.[9]

Using 1992-2013 information from the Surveillance, Epidemiology, and End Results (SEER) Program database, Challapalli et al looked at the disease-specific survival rates in the United States for adult blacks, Hispanics, Asians/Pacific Islanders, and American Indians/Alaska natives, with NPC, finding the worst disease-specific survival rate to be among non-Hispanic American Indians/Alaska natives. The best disease-specific survival rate was found in non-Hispanic Asians/Pacific Islanders.[10]

Etiology

A clear etiology for nasopharyngeal carcinoma (NPC) is still lacking. In general, NPC is thought to be the result of both genetic susceptibility and environmental factors such as carcinogens and infection with Epstein-Barr virus (EBV). Evidence in support of genetic factors is the association of NPC with genotypes HLA-A2 and HLA-Bsin2, which are prevalent in individuals from southern China but rare in whites. Furthermore, abnormalities of multiple chromosomes, including 1, 2, 3, 4, 5, 6, 8, 9, 11, 13, 14, 15, 16, 17, 22, and X, have been identified.

Possible environmental or cultural factors that may be associated with NPC include the ingestion of Cantonese-style salted fish and preserved foods that contain carcinogenic nitrosamines, especially during childhood. Evidence of EBV-DNA in almost all NPC cells that were studied supports the association of NPC with EBV. Further, the detection of clonal EBV-DNA in NPC suggests that the malignancy is a clonal expansion of a single EBV-infected progenitor cell. This finding indicates that EBV is present within the cell at the time of malignant transformation and suggests a role for the virus in contributing to the early transformation event. The contribution of both genetic factors and environmental factors for this disease is reflected in the observation that the incidence of NPC for American-born, second-generation Chinese individuals is lower than that for Chinese-born individuals in China but remains higher than that for white individuals in the United States.

Presentation

Although reported in all age groups, a bimodal peak incidence appears to occur in individuals aged 30-40 years and 50-60 years. Nasopharyngeal carcinoma (NPC) is observed predominantly in males, with a male-to-female ratio of 3:1. Clinically, NPC has few early warning signs, which often means late diagnosis. Early but nonspecific symptoms include nasal obstruction, blood-tinged sputum or nasal discharge, tinnitus, headache, ear fullness, and unilateral conductive hearing loss from serous otitis media or recurrent acute otitis media. In advanced cases, the tumor can invade the skull base and spread intracranially through one of the many nearby foramina. Evidence of cranial nerve involvement (III-VI), including diplopia and numbness of the face, suggests cavernous sinus invasion.

The abundant supply of regional lymphatic vessels in the nasopharynx contributes to the high prevalence of cervical metastasis. Approximately 44-57% of patients initially seek medical attention because of a metastatic lymph node that manifests as a neck mass. At the time of diagnosis, 60-85% of patients already have cervical metastasis.

Systemic dissemination also occurs more readily in NPC than in other head and neck cancers. The most frequently involved sites are bone, lung, and liver. Distant metastases are present in 5-10% of patients at the initial presentation.

Indications

Salvage nasopharyngectomy and neck dissection may be indicated in patients with nasopharyngeal carcinoma (NPC) that persisted or recurred locoregionally following prior treatment with radiation with or without chemotherapy. The proper selection of patients and surgical approach are essential for a successful outcome.

Relevant Anatomy

Nasopharynx

The nasopharynx is defined anteriorly by the posterior choanae, posteriorly by the clivus and the first 2 cervical vertebrae, superiorly by the floor of the sphenoid, and inferiorly by the level of the free border of the soft palate. The nasopharynx is divided into 3 subsites: the posterosuperior wall, the lateral walls, and the posterosuperior surface of the soft palate. The torus tubarius is the opening of the eustachian tube into the lateral nasopharyngeal wall. The fossa of Rosenmüller is the groove or recess posterior to the torus at the junction between the lateral and posterior walls. Nasopharyngeal carcinoma (NPC) most commonly occurs in this location.

The posterior and lateral nasopharyngeal walls are composed of 3 layers of tissue. The mucosal epithelium of the nasopharynx is complex, consisting mainly of pseudostratified columnar ciliated epithelium near the choanae and the adjacent part of the roof of the nasopharynx, a transitional epithelium in the roof and the lateral walls, and stratified squamous epithelium along the posterior and inferior portions of the nasopharynx. The superior constrictor muscle and the buccopharyngeal fascia surround the mucosa. Superiorly, the buccopharyngeal fascia unites with the pharyngobasilar fascia, which is attached to the skull base.

The buccopharyngeal fascia extends posterolaterally from the free edge of the medial pterygoid plate to the lateral border of the carotid artery. This fascia separates the nasopharynx from the parapharyngeal (paranasopharyngeal) space. A line joining the free edge of the medial pterygoid plate posterolaterally to the styloid process divides the paranasopharyngeal space into the prestyloid space anteriorly and the retrostyloid space (containing the carotid sheath and the cranial nerves) posteriorly.

The paranasopharyngeal space is bound anteriorly by the pterygomandibular raphe, which joins the lateral pterygoid plate to the mandible. The retropharyngeal space contains the retropharyngeal lymph nodes and the node of Rouviere. This space is located posterior to the buccopharyngeal fascia and anterior to the prevertebral fascia; therefore, lesions that extend beyond the buccopharyngeal fascia posteriorly involve the retropharyngeal space, while lesions extending laterally beyond this fascia reach the parapharyngeal space.

The nasopharynx is an anatomically difficult area to expose surgically. This area is in close proximity to several foramina and associated vital neurovascular structures. These include the foramen ovale, the foramen spinosum, the foramen lacerum, the carotid canal, and the jugular foramen.

Neck

Ho originally described the supraclavicular fossa as a triangular region defined by 3 points: the sternal end of the clavicle, the lateral end of the clavicle, and the point where the neck meets the shoulder.[11] This area is clinically significant in that any nodal involvement within this triangle is, by definition, an N3 lesion and, therefore, stage IV cancer.

Contraindications

Salvage nasopharyngectomy is contraindicated in patients with locally unresectable recurrent nasopharyngeal cancer and patients with distant metastasis.

Laboratory Studies

Many studies have shown that nasopharyngeal carcinoma (NPC) is closely associated with EBV. Elevated EBV titers may also be associated with other disease entities, such as sinonasal undifferentiated carcinoma (SNUC), sinonasal lymphoma, and tongue cancer.

Seroepidemiologic studies have demonstrated that 80-90% of patients with World Health Organization (WHO) type 2 NPC and WHO type 3 NPC have elevated levels of immunoglobulin A (IgA) antibodies to viral capsid antigen (VCA) and early antigen (EA). However, only 10-20% of patients with WHO type 1 NPC have elevated levels of IgA to VCA.

Low et al examined the EBV serology in 111 patients with NPC and in 111 healthy patients.[12] In the patients with NPC, 80.2% tested positive for IgA to EA, and 97.3% tested positive for IgA to VCA. In the control group, 100% tested negative for IgA to EA, but only 46.8% tested negative for IgA to VCA. In other words, the positive predictive value (PPV) of the EA serology is 100%, while the negative predictive value (NPV) is 83.5%. For VCA serology, the PPV is 64.7%, while the NPV is 94.5%. Therefore, a patient who tested positive for EA serology has a 100% chance of having NPC. A patient who has negative VCA serology only has a 5.5% chance of having NPC. A difficult clinical situation arises if a patient has a negative EA serology test but has a positive VCA serology. This serology combination predicts a 37.8% chance of having NPC. See Tables 1-2.

Table 1. Diagnostic profiles of IgA to EA and IgA to VCA[12]



View Table

See Table

 

Table 2. Predictive Value of Epstein-Barr Virus Serology Combinations



View Table

See Table

 

Other serologic tests include IgA antibodies directed against EBV, EBV nuclear antigen (EBNA)–1 (found in about 90% of patients with NPC), and immunoglobulin G (IgG) antibodies to the EBV replication activator (ZEBRA) and BRLF1 transcription activator (Rta). Cai et al[13] investigated the diagnostic significance of 4 EBV antibodies in 211 NPC patients and 413 healthy controls and found similar sensitivity and specificity for IgA to EA and IgA to VCA (Table 3), as described by Low et al. (Table 2). They then determined the diagnostic accuracy based on different combinations of these 4 EBV antibody titers (Table 4).

The utility of using a combination of IgA to VCA and IgA to EBNA-1 as a screening tool in an endemic area of Southern China was reported by Liu et al.[14] In this study, 28,688 patients were screened and 862 patients were found to have positive serology. These patients underwent further diagnostic evaluation including fiberoptic nasopharyngoscopy and nasopharyngeal biopsy. Of these, 38 (4.4%) were eventually diagnosed with NPC, the majority of which was at early stages.

Table 3. Diagnostic Values of the 4 EBV Antibodies[13]



View Table

See Table

 

Table 4. Diagnostic Accuracy based on Different Combination of the 4 EBV Antibodies



View Table

See Table

 

Recently, several authors also reported on the potential use of plasma EBV DNA as a screening tool either alone or in combination with other serological EBV antibodies.[15, 16]

Imaging Studies

Assessment of locoregional disease

MRI with gadolinium and fat suppression is the radiologic modality of choice.[17] Determine if any intracranial extension of the tumor involves the brain parenchyma or the cavernous sinus. Intracranial spread can occur through several foramina that are in close proximity to the nasopharynx. These foramina include the foramen ovale, the foramen spinosum, the foramen lacerum, the carotid canal, and the jugular foramen.

Detect any tumor extension into the retropharyngeal, parapharyngeal, and pterygomaxillary spaces, as well as the infratemporal fossa and the sinuses.

Workup for metastatic disease

In a study involving 150 patients with untreated nasopharyngeal carcinoma (NPC), whole-body MRI and18 F-FDG positron emission tomography (PET)/CT showed similar diagnostic accuracy of 90.5% and 87.8% respectively in assessing distant site metastasis.[18]

In another study involving 78 patients, PET-CT scan was shown to be more sensitive and specific than CT scan in assessing for distant metastasis to lungs, liver, and bones.[19]

Detection of residual and/or recurrent nasopharyngeal carcinoma following definitive radiation with or without chemotherapy

Detection of residual and/or recurrent NPC following radiation treatment can be challenging because of radiation-induced scarring, fibrosis, and edema of the nasopharyngeal tissue. This treatment-related anatomic distortion may limit the role of anatomic-based imaging modalities such as MRI and CT. Function-based imaging modalities, such as18 F-FDG PET scan, are not affected by this anatomic distortion. In a systemic review of 21 articles, Liu et al found that18 F-FDG PET scan has an average sensitivity 95% in detection of local residual or recurrent disease, which is significantly higher compared with CT scan (76%) and MRI (78%).[20]

Function-based imaging modality, however, also has its limitations in cancer detection. The fact that both the cancer and the inflammatory tissues take up18 F-FDG limits the ability of the18 F-FDG PET-CT scan to distinguish viable tumor tissue from radionecrosis or osteomyelitis, resulting in high false-positive interpretation. Furthermore, because the hypermetabolic brain tissue provided a high background, recurrent/residual tumor located intracranially may be missed by18 F-FDG PET-CT scan, resulting in a high false-negative rate.

Another function-based imaging,201 TI single photon emission computed tomography (SPECT)/CT scan is based on thallium-201, which is a potassium analogue that competes with potassium for intracellular transport across the cell membrane via sodium-potassium-ATPase pump system in tumor cell membrane.[21] Because the necrotic tissue cell membrane lack the sodium-potassium-ATPase pump,201 TI does not accumulate in areas of osteoradionecrosis, and its uptake reflects the presence of viable tumor. This modality, however, is limited by spatial resolution and can miss tumors less than 1.5 cm in size.[22]

In a study comparing the use of201 TI SPECT/CT versus18 F-FDG PET-CT in detecting recurrent skull base nasopharyngeal carcinoma, Yen et al found the accuracy of these 2 modalities to be similar. The sensitivity and specificity for201 TI SPECT/CT were 66.7% and 100%, and those for18 F-FDG PET-CT were 86.7% and 75%.[22]



View Image

Axial T2-weighted image shows a left-sided cervical nodal metastasis resulting from nasopharyngeal cancer.

Diagnostic Procedures

See the list below:

Histologic Findings

Nasopharyngeal carcinoma (NPC) can be grouped into the following 3 categories according to the WHO classification system:

Staging

The American Joint Committee on Cancer-Union Internationale Contre le Cancer (AJCC-UICC) 2002 Classification is as follows:

Medical Therapy

Previously untreated nasopharyngeal carcinoma

Nasopharynx

External beam radiation therapy is the primary mode of management of nasopharyngeal carcinoma (NPC), both at the primary site and in the neck. This is mainly because of this tumor's high degree of sensitivity to radiation as well as the anatomical constraints for surgical access to the highly complex nasopharyngeal region. Recent advances in imaging capabilities (eg, the ability to more accurately define tumor location) and improved radiotherapy techniques (eg, stereotactic radiotherapy boost) have helped to improve the locoregional control rate. At the same time, complications associated with radiation therapy have been reduced. Various sophisticated fractionation schema and boosting techniques have been advocated, with a minimum of 65-75 Gy of radiation delivered to the primary site.

Although radiation therapy alone is a well-accepted management of stages I and II NPC, the administration of chemotherapy adjunctive to radiotherapy in advanced NPC (stages III-IV) has remained a controversial issue because of conflicting reports in the literature. When interpreting these data, the inherent difference in disease type in endemic areas (eg, Southeast Asia) and nonendemic areas (eg, North America and Western Europe) must be noted. WHO type 1 makes up fewer than 5% of all NPC cases in Southeast Asia, while it accounts for 25% of tumor types in the Intergroup Study from North America.[30] WHO type 1 NPC is generally considered to be less radiosensitive than WHO type 2 and 3 NPC; therefore, WHO type 1 NPC is associated with the worst prognosis. Not surprisingly then, the locoregional control and survival rates reported from Southeast Asia are, in general, better than rates reported from the West.

Another important thing to remember is that the radiation regimens reported in Asian studies are typically more aggressive than ones reported in nonendemic areas in the West.[31] This may account for the better outcome in the radiation-alone arm seen in Asian studies, making it harder to show survival benefit with the addition of chemotherapy. Finally, many different types of chemotherapeutic regimens (different drugs, concentrations, and different timing) were used in these trials, which may also account for some of the differences in outcome.

Chemotherapy can be delivered before (neoadjuvant), during (concurrent), or following (adjuvant) radiation therapy. Active chemotherapeutic agents include cisplatin, 5-fluorouracil (5-FU), doxorubicin, epirubicin, bleomycin, mitoxantrone, methotrexate, and vinca alkaloids. Various chemotherapeutic approaches have been devised to improve the response rates while minimizing toxicities.

In 1998, a landmark study (Intergroup Study 0099) was reported by Al-Saraaf et al.[30] This was a large, prospective, randomized trial from North America that demonstrated that concomitant chemoradiation (cisplatin 100 mg/m2 infused on days 1, 22, and 43) followed by adjuvant chemotherapy with cisplatin (80 mg/m2) and 5-FU (1 g/m2) every 4 weeks for 3 cycles improved overall survival (OS) at 3 years for patients with advanced stages of NPC over radiation therapy alone (75% vs 46%).

In an updated report, Al-Saraaf reported that patients treated with chemoradiation continued to have superior OS rates over patients treated with radiation alone at 5 years (67% vs 37%).[32] This study was the first large randomized trial that demonstrated a significant improvement in OS with the addition of chemotherapy to radiation. Following this landmark study, a major change in the treatment paradigm for patients with advanced stage NPC occurred in the United States. Most centers now treat patients who have advanced-stage NPC with a combination of chemotherapy and radiation. However, this treatment regimen is certainly not routinely practiced in other parts of the world, especially in endemic regions in Asia.

In contrast to the Intergroup Study 0099, several large, prospective, randomized trials failed to show any survival benefit with the addition of chemotherapy for treatment of advanced NPC (see Table 5 below). Most of these earlier studies from Asia, however, used chemotherapy either in the neoadjuvant or adjuvant fashion instead of the concurrent and adjuvant regimen used in Intergroup Study. Using chemotherapy in an adjuvant setting, the Taiwan Cooperative Oncology Group Trial failed to demonstrate any survival benefit from the addition of adjuvant chemotherapy in advanced stages of NPC. The 5-year OS for the chemoradiation group was 61% versus 55% survival for radiation alone group.[33]

Studies using chemotherapy in a neoadjuvant setting also failed to show survival advantage.[34] A randomized trial from the Asian-Oceanian Clinical Oncology Association compared induction chemotherapy followed by radiotherapy versus radiotherapy alone.[35] The 3-year OS rate was 78% for the induction chemotherapy group, which was not significantly different from the 3-year OS of 71% for the radiation alone group.

In a similar large, randomized trial, Ma et al reported a 5-year OS of 63% for the group that received induction chemotherapy followed by radiation versus a 5-year OS of 56% for the group that underwent radiation alone.[36] Statistically significant improvement in survival was not achieved (P =0.11). Finally, Chan et al evaluated the addition of both neoadjuvant and adjuvant chemotherapy and reported a 5-year OS rate of 80% for the chemoradiation group and 81% for the radiation alone group with P =0.1.[37]

Unfortunately, the disparate results in these well-conducted large, prospective, randomized trials are difficult to interpret, for several reasons. First, the difference in the proportion of the 3 types of NPC in each of these studies may contribute to some of the disparities in the effect of chemotherapy in these different studies. As stated previously, most of the NPC cases (>95%) in Southeast Asia involve type 2 NPC and type 3 NPC, which are extremely radiosensitive. Treatment with radiation alone usually results in an excellent 5-year OS rate, ranging from 60-80%. Improving on this excellent survival rate is difficult with the addition of chemotherapy. Although fewer than 5% of patients in these studies from Southeast Asia have type 1 NPC, a large proportion of patients (25%) in the Intergroup Study have type 1 NPC, which is less radiosensitive and is associated with a much lower survival rate than radiation alone.[30]

The possibility exists that the significant improvement in survival reported from the Intergroup Study may be more applicable to patients with type 1 NPC than patients with type 2 or 3 NPC.[30] Subgroup analysis of the Intergroup Study seems to support the significant beneficial effect of adding chemotherapy for patients with WHO type I NPC (see Table 6 below). In patients with WHO type I treated by radiation alone, the 5-year OS rate is only 14%. The survival rate increased to 59% in the same group of patients treated with chemoradiation. However, one must be cautious in drawing conclusion from subset analysis, especially given the small number of patients in this subset of WHO type I (n = 36).

The debate over whether chemotherapy is beneficial in the treatment of advanced NPC is further complicated because different trials used different chemotherapeutic agents (eg, vincristine, cisplatin, bleomycin, epirubicin, 5-FU, methotrexate) as well as different delivery schedules (ie, neoadjuvant, concurrent, adjuvant, combination). The improved survival rate from the Intergroup Study may result from the concurrent use of chemotherapy with radiation, whereas many prior studies from Asian countries mainly use chemotherapy in the adjuvant setting, neoadjuvant setting, or both.

To test this hypothesis, several phase III randomized trials were conducted in Asia using chemotherapy in the concurrent fashion with radiotherapy. Unfortunately, the results were again inconclusive. Chi et al reported that the concurrent use of cisplatin, 5-FU, and leucovorin with radiation failed to improve 5-year overall survival in stage IV NPC patients when compared with radiation alone (61% versus 55%). On the other hand, Chan et al reported on a phase III randomized study comparing concurrent use of weekly infusion of cisplatin (40 mg/m2) during radiation versus radiation alone.[37] They demonstrated that for patients with stage II and III NPC, the 5-year OS rate is better in patients treated with concurrent chemoradiotherapy (70.3%) than for patients treated with radiation alone (58.6%).

Another study from Taiwan reported by Lin et al also demonstrated that the use of concurrent chemoradiotherapy is superior to radiotherapy alone.[38] In that study, patients were treated with 2 cycles of cisplatin and 5-FU during radiation. The 5-year OS rate in the chemoradiation group (72.3%) was significantly better than that for the radiation alone group (54.2%).

Two published randomized phase III clinical trials, using the exact same treatment schema that was used in the Intergroup Study 0099, however, gave contradictory results. Using concurrent chemoradiation with cisplatin followed by adjuvant chemotherapy with cisplatin and 5-FU, Wee et al reported a statistically significant (P =0.008) improvement in 5-year OS rate in patients who received chemoradiotherapy (67%) versus patients who received radiation alone (49%).[39, 40] However, using this same regimen, Lee et al reported no statistically significant difference (P =0.22) in 5-year OS rate in patients treated with chemoradiotherapy (68%) versus patients treated with radiation alone (64%) (see Table 5 below).[41, 42] Nonetheless, the locoregional control rate in the chemoradiation group (92%) is statistically significantly (P =0.027) better than that for the radiation alone group (82%).

Further, this was the first randomized trial to include a more contemporary standard of radiation, with approximately 60% of patients receiving some form of intensity-modulated radiotherapy (IMRT) or 3-dimensional conformal radiotherapy (3D-CRT). As stated earlier, radiation regimens used in Asian studies are typically more aggressive and treatment with radiation alone usually results in an excellent 5-year OS rate. Thus, it may be more difficult to improve on this excellent survival rate (using radiation alone) with the addition of chemotherapy.

Finally, a recently published randomized phase III trial from Chen et al involved 316 patients from Chinese mainland.[43, 44] Patients in the combined treatment arm were given concurrent cisplatin (40 mg/m2) weekly during 7 weeks of radiation followed by cisplatin (80 mg/m2 on day 1) and fluorouracil (800 mg/ m2 on days 1-5) every 4 weeks for 3 cycles after radiation. The 5-year OS for the chemoradiation arm and radiation-alone arm were 72% and 62%, respectively (hazard ratio, 0.69; 95% CI, 0.48-0.99; P =0.043.[44]

A meta-analysis involving 1,528 patients from 6 randomized studies comparing combined chemotherapy-radiation therapy versus radiation therapy alone in locally advanced NCP showed that the addition of chemotherapy to radiation therapy increased disease-free/progression-free survival by 37% at 2 years, 40% at 3 years, and 34% at 4 years after treatment. Likewise, the OS increased by 20% at 2 years, 19% at 3 years, and 21% at 4 years with chemotherapy plus radiation therapy.[45]

A report from the Meta-Analysis of Chemotherapy in Nasopharyngeal Carcinoma (MAC-NPC) reviewed individual patient data from 8 well-designed randomized trials that compared chemotherapy plus radiotherapy versus radiotherapy alone in locally advanced NPC.[46] A total of 1753 patients were included in this review. All trials used conventional radiotherapy and cisplatin-based chemotherapy. The authors found that the addition of chemotherapy improved 5-year OS from 56% to 62% (absolute survival benefit, 6%) and improved EFS from 42% to 52% (absolute benefit, 10%).

They also observed a significant interaction between chemotherapy timings and OS (P =.005), which explained the heterogeneity of clinical trial results previously noted. The use of concurrent chemotherapy with radiation was found to result in the highest survival benefit. In the sensitivity analysis, chemotherapy was found to be more efficient against WHO type 1 disease than other types. The authors concluded that the addition of chemotherapy to standard radiotherapy provides a small but significant survival benefit in patients with nasopharyngeal carcinoma. This benefit is especially observed when chemotherapy is administered concomitantly with radiotherapy. The role of induction chemotherapy and adjuvant chemotherapy is more questionable.

A recent large, phase 3, multicenter, randomized control trial[47] compared the effect of the addition of adjuvant chemotherapy (cisplatin and 5-FU) to concurrent chemotherapy with cisplatin in locoregionally advanced NPC. This trial involved 7 institutions in China and enrolled patients with stage III or IV NPC, except for T3-4N0. A total of 251 patients were randomized to the concurrent chemoradiation plus adjuvant chemotherapy (C+A) group, while 257 were randomized to the concurrent chemoradiation alone (C) group. After a median follow-up of 37.8 months, the estimated 2 year failure-free survival rate was 86% (95% CI, 81-90) in the C+A group and 84% (95% CI, 78-88) in the C group (hazard ratio 0.74; 95% CI, 0.49-1.10; P =0.13).

Similarly, in a meta-analysis of 5 studies involving 394 patients in the C+A group and 399 patients in the C only group, Liang et al showed that the addition of adjuvant chemotherapy to concurrent chemoradiation did not improve outcome compared with concurrent chemoradiation alone. Risk ratios of 1.02 (95% CI, 0.89-1.15), 0.93 (95% CI, 0.72-1.21), 1.07 (95% CI, 0.87-1.32), and 0.95 (95% CI, 0.80-1.13) were observed for 3 years OS, 5 years failure-free survival, 5 years locoregional failure-free survival, and 5 years distant metastasis failure-free survival, respectively.[48]

In his recent review paper, Al-Sarraf[32] commented on reversing the sequence of chemotherapy from adjuvant to neoadjuvant. Using this protocol of induction chemotherapy with 3 courses of cisplatin and 5-FU followed by concurrent chemoradiation using 3 courses of cisplatin, he reported an unpublished 5 year OS of approximately 90%. Kong et al[49] recently reported on the use of neoadjuvant chemotherapy consisting of a taxane, cisplatin, and 5-FU (TPF regimen) followed by concurrent chemoradiation in 52 stage III and 64 stage IVA/IVB NPC patients. The 3-year OS was 94.8% (95% CI, 87.6-100%) for stage III patients and 90.2% (95% CI, 81.8-98.6%) for stage IVA/IVB patients. This excellent result is very encouraging and warrants randomized controlled trials.

Table 5. Prospective Randomized Clinical Trials of Chemoradiation Versus Radiation Alone in the Treatment of Locally Advanced NPC



View Table

See Table

Table 6. Intergroup Study 0099. Subgroup Analysis of 5-Year Overall Survival Based on WHO Types[30]



View Table

See Table

Neck

Radiation therapy more readily controls neck disease that arises from NPC than comparable neck disease from other head and neck carcinomas. Regional control remains a possibility even with extensive nodal disease. Delivery of radiation at a minimum of 65-75 Gy to the clinically positive neck node is recommended. Given the high propensity of NPC to metastasize to the neck, most authors recommend elective treatment of the N0 neck. Furthermore, treatment of both sides of the neck is recommended. The retropharyngeal and parapharyngeal lymph nodes are included in the treatment volume of the primary tumor.

Persistent or recurrent nasopharyngeal carcinoma (locoregional failures)

Despite recent advances in the management of NPC, locoregional failure is still significant, with reported rates of 15.6-58% (median, 34%).[4, 5, 6, 7]

Nasopharynx - Local failure

The frequency of local failure is reported to range from 18-58%.[54, 55] Management of locally recurrent diseases can be accomplished with either re-irradiation or salvage nasopharyngectomy. Re-irradiation is associated with a high frequency of complications, including temporal lobe necrosis, brainstem damage, cranial neuropathy, endocrine dysfunction, visual and hearing impairments, osteonecrosis, soft tissue necrosis, and trismus. These complications can be reduced with the use of brachytherapy or stereotactic radiotherapy. Although the potential for these complications is high, only 10-30% of patients achieve local control after the second course of irradiation.[56, 57] A survival rate of 34-48% at 3 years has been reported, although only about 15-23% of patients achieve disease-free survival.[56, 57]

Neck - Regional failure

The frequency of persistent or recurrent neck disease is reported to range from 8-34%.[58] Patients whose treatment failed regionally can be treated with either re-irradiation or salvage neck dissection. The control rate after re-irradiation is reported to be between 28% and 33%.[57] In contrast, Wei et al reported a regional control rate of 66% after radical neck dissection.[59] Despite a relatively good chance of regional control, these patients who presented with recurrent or persistent neck disease usually have a high risk of distant metastases.

Distant metastases

A high prevalence of distant metastases has been observed for patients with NPC, with a substantial number eventually experiencing distant failure despite lasting locoregional control. The distant failure rate was reported to range from 18-35%. At the time of initial presentation, 5-10% of patients may already have distant metastases. The occurrence of distant disease does not appear to be associated with the size of the primary tumor. However, a strong association exists between nodal disease and the development of distant disease, with 38% of patients with N+ neck disease exhibiting distant metastases versus 11% of patients with N0 neck disease. Some series have reported a rate of up to 80% of distant metastasis in patients with N3 neck disease.

The lung is the most common site of metastasis, followed by bone and the liver. Currently, available treatment modalities are ineffective in curing distant metastases. Palliative treatment is directed toward pain relief, symptom control, and prolongation of life. Radiation can be extremely effective in palliating pain from bone metastasis. Although the use of palliative chemotherapy in a patient who experiences symptoms is reasonable, the use of palliative chemotherapy in a patient without any symptoms is not as clear. The desire for prolongation of life must be balanced against the patient's quality of life, which should be the first priority.

Surgical Therapy

Previously untreated nasopharyngeal carcinoma

Because of the tumor's high degree of sensitivity to radiation and the anatomical constraints for surgical access to the highly complex nasopharyngeal region, nasopharyngectomy is reserved only for treatment of recurrent NPC with limited disease.

Persistent or recurrent nasopharyngeal carcinoma (locoregional failures)

Nasopharynx - Local failure

Although nasopharyngectomy can achieve slightly better local control and a lower rate of complication as compared with re-irradiation, this surgery is only applicable in patients with limited disease such as rT1, rT2, rT3. A recurrent or residual disease that involves the middle cranial fossa may be amenable to resection through the craniofacial approach. Most surgeons consider the involvement of the cavernous sinus, the cranial nerves, and the carotid artery as a contraindication for surgical intervention. Even though tumors in these areas are technically resectable, salvage surgery is not advised because of the high morbidity associated with resection of the internal carotid artery and the cranial nerves in the setting of a very low probability of cure. A recent meta-analysis of 779 patients from 17 published studies showed a 5-year overall survival of 51.2% following salvage surgery with or without reirradiation for recurrent nasopharyngeal carcinoma.[60]

Various surgical approaches to the nasopharyngeal region have been described. Each approach has its own merit, and no single approach has clearly been shown to be superior to the other approaches. Because of the nature of the disease process, which involves an extremely complex anatomical region, the surgeon needs to be familiar with all of these surgical approaches. The operation performed must be tailored to the areas involved by the tumor and may involve a combination of approaches, thus allowing maximal exposure while minimizing associated morbidity.

Neck - Regional failure

Radical neck dissection can be used to treat recurrent or residual disease in the neck after radiation treatment with a good probability of regional control. Wei et al reported a regional control rate of 66% after radical neck dissection.[59] On serial sectioning of the entire radical neck dissection specimen, 27.5% of the specimen was found to have tumors lying in close proximity to, or even infiltrating, the spinal accessory nerve. Based on this finding, Wei et al recommended radical neck dissection as the salvage procedure of choice.[59] If the retropharyngeal or parapharyngeal space is involved, neck dissection is extended to include this region.

Preoperative Details

A detailed assessment of the extent of tumor involvement is extremely important. Most surgeons consider the involvement of the cavernous sinus as a contraindication for surgery. A clear appreciation of the tumor in relation to the internal carotid artery is essential. Metastatic workup must be performed to exclude distant metastases.

Intraoperative Details

Fee describes a transpalatal, transmaxillary, and transcervical approach.[24] This approach to the nasopharynx provides excellent exposure to both sides of the nasopharynx with minimal morbidity to the patient. Isolation and protection of the internal carotid artery through the transcervical approach allow resection of the lateral nasopharyngeal wall with minimal risk to the internal carotid artery and the cranial nerves. Disease that extends to the pterygomaxillary space can be exposed via a transmaxillary approach through the posterior wall of the maxillary sinus. Then, the clivus and vertebral body bone are drilled with a large cutting burr. Fee reported on his experience with 33 patients who had recurrent NPC and were monitored for 2-17 years after nasopharyngectomy. A 5-year local control rate of 67% with a 5-year disease-free survival rate of 52% and an OS rate of 60% were achieved.

Fisch describes the infratemporal fossa approach, and Gross and Panje describe the lateral temporal approach.[61, 62] Both approaches provide excellent exposure of tumors that extend into the infratemporal fossa and the parapharyngeal space. A major disadvantage of these approaches is that entry into the nasopharynx is performed on the side of the lesion, making complete excision difficult if the tumor extends to the contralateral nasopharynx. Furthermore, the morbidity following this approach is significant and may include sensorineural hearing loss, cerebrospinal fluid (CSF) leak, unilateral laryngeal paralysis, and facial nerve deficit.

Wei et al suggested a new idea for exposure of the nasopharynx through the maxillary swing (facial translocation).[63] This approach involves a Weber-Fergusson incision. After achieving the necessary bone cuts, the entire osteocutaneous complex is swung laterally to provide exposure of disease in the ipsilateralpterygomaxillary and paranasopharyngeal space. However, the control of the internal carotid artery is less than optimal. Wei reported a local control rate of 42% at 3.5 years.[63]

Biller and Krespi describe the transcervico-mandibulo-palatal approach. This approach provides a wide-field exposure of the nasopharynx and excellent protection of the internal carotid artery. Morton et al reported a 67% local control rate at 2 years with this approach.[64] King et al reported on a series of 31 patients who were treated with a variety of surgical approaches followed by postoperative radiation.[65] They reported a 5-year survival rate of 47% with a 5-year disease-free survival rate of 42%.

With the increasing interest in minimally invasive surgery in the field of head and neck, endoscopic approach for resection of recurrent nasopharyngeal carcinoma has been described. Chen et al. reported on a series of 6 patients with recurrent T1 or T2a disease who underwent endoscopic nasopharyngectomy with a local control rate of 83% at a mean follow-up duration of 29 months.[25]

Finally, the introduction of the da Vinci robot provided another technological advancement that enables surgeons to reach and operate in areas such as the nasopharynx, which are difficult to access. Tsang et al[66] reported on 12 patients who underwent robotic nasopharyngectomy with 2-year local control rate of 86% and 2-year OS of 83%.

Postoperative Details

The nasopharyngeal defect is covered with a split-thickness skin graft. The graft is held in place with packing. A 14F Foley catheter is then placed through the nose, and the balloon is inflated to keep the packing in place. Bilateral or unilateral myringotomy and tube placement are performed at the end of the surgery. The Foley catheter is usually removed on the third postoperative day, and the nasopharyngeal packing is removed on the 10th postoperative day. The patient is instructed to irrigate the nasopharynx with normal saline until healing is complete.

Follow-up

Unlike other head and neck cancers, NPC is known for its continued risk of late recurrences, and long-term follow-up care is required. Although most recurrences occur within 5 years, 5-15% of recurrences may manifest between the 5th and 10th year. Therefore, patients with NPC should be monitored for at least 10 years after treatment. Some authors have suggested that a 10-year, rather than the 5-year, survival rate is needed to assess the effectiveness of a particular treatment of NPC.

Complications

Radiation

Recent advances in imaging capabilities (which more accurately define tumor volumes) and improved radiotherapy have helped in improving the locoregional control rate while, at the same time, reducing the complications associated with radiation therapy. However, in an attempt to improve locoregional control and survival rates, a higher radiation dose, a more radical fractionation schedule, and the addition of chemotherapy have, in some cases, increased the frequency and severity of complications.

Complications associated with radiation therapy to the nasopharynx and the neck can be classified according to the following organ systems:

Although intensity-modulated radiation therapy (IMRT) is considered an improved means of radiation delivery, a cross-sectional cohort study by McDowell et al found that patients with NPC who underwent successful IMRT nonetheless suffered symptoms that impacted quality of life even years after treatment. Difficulties encountered frequently included hearing problems and also involved difficulties with dry mouth, mucus, swallowing/chewing, memory, and the teeth/gums. Moreover, hypothyroidism developed in 69% of the cohort. Patients demonstrated depression, anxiety, and fatigue rates of, respectively, 25%, 37%, and 28%, and these were significantly associated with quality of life.[69]

 

Surgery

Surgical complications can be divided into those associated with nasopharyngectomy and those associated with neck dissection. Because surgery is usually performed after a course of radical radiotherapy, complications from poor wound healing are commonly observed. These complications include palatal fistula, nasopharyngeal wound infection, osteonecrosis, osteomyelitis of cervical vertebrae or skull base, nonunion or malunion of osteotomy sites, and wound edge or flap necrosis. Other complications include damage to the internal carotid artery or the cranial nerves, dural violation at the skull base, and death.

Outcome and Prognosis

The prognostic factors for patients with nasopharyngeal carcinoma (NPC) include the extent of the primary tumor (ie, skull base invasion, cranial nerve involvement, parapharyngeal infiltration), the level of the disease in the neck, the histologic subtype, the age and the sex of the patient, and the type and technique of radiotherapy. Survival rates are generally better in females than in males.

Some of the largest studies have reported a 5-year disease-free survival rate of 40-60% with primary radiation treatment. The 5-year overall survival (OS) rate is 85-95% for stage I NPC and 70-80% for stage II NPC treated with radiation alone. For stages III and IV NPC treated with radiation alone, the 5-year OS rate ranges from 24-80%, with better results generally occurring in patients from Southeast Asia. The Intergroup Study 0099 demonstrated that North American patients with advanced NPC benefited from concurrent chemotherapy with an improved 5-year OS rate of 67% compared with the 5-year OS rate of 37% for patients treated with radiation alone.

WHO type 3 NPC or undifferentiated carcinoma has the most favorable prognosis because of its high degree of radiosensitivity. The 5-year OS rate is 60-80%. In contrast, WHO type 1 NPC has the worst prognosis, with a 5-year OS rate of 20-40% because of its low radiosensitivity.

Unlike other head and neck carcinomas, some NPCs have a long, protracted course. Some patients can live with their recurrent disease for many years before succumbing to the disorder.

A literature review by Liao et al indicated that in patients with NPC, overexpression of matrix metalloproteinase-9 correlates with a poor prognosis for overall survival and disease-free survival.[70]

Future and Controversies

Several areas continue to be debated regarding the management of nasopharyngeal carcinoma (NPC).

The role of chemotherapy in advanced nasopharyngeal carcinoma

Unfortunately, the literature is conflicting regarding the role of chemotherapy in the management of advanced NPC. This discrepancy in the literature may result from differences in the proportion of NPC WHO types, the types of chemotherapeutic agents, and delivery schedules in these various clinical trials. The significant improvement in survival with the addition of chemotherapy reported from the Intergroup Study may be because of the large proportion of patients with type 1 NPC in this study and the concurrent use of chemotherapy. Other large clinical trials, most notably from Asia, include a large proportion of patients with type 2 or 3 NPC who received chemotherapy in the neoadjuvant or adjuvant fashion. These trials failed to demonstrate improvement in overall survival (OS) with the addition of chemotherapy.

Several clinical trials from Asia that incorporated the use of concurrent chemoradiotherapy for locoregionally advanced NPC did show statistically significant improvement in OS. However, results are still conflicting. Using the same regimen as the one used in Intergroup Study 0099, Lee et al reported no statistically significant difference in 3-year OS in patients treated with chemoradiotherapy (76%) versus patients treated with radiation alone (77%). Nonetheless, the locoregional control rate in the chemoradiation group (93%) is statistically significantly better than the radiation alone group (82%).

Most recently, a report from the Meta-Analysis of Chemotherapy in Nasopharyngeal Carcinoma (MAC-NPC) reviewed individual patient data from 8 well-designed, randomized trials comparing chemotherapy plus radiotherapy with radiotherapy alone in locally advanced NPC. A total of 1753 patients were included in this review. The authors found that the addition of chemotherapy improved 5-year OS from 56% to 62% (absolute survival benefit, 6%) and improved EFS from 42% to 52% (absolute benefit, 10%). The authors concluded that the addition of chemotherapy to standard radiotherapy provides a small but significant survival benefit in patients with nasopharyngeal carcinoma. This benefit is essentially observed when chemotherapy is administered concomitantly with radiotherapy. The role of induction chemotherapy and adjuvant chemotherapy is more questionable.

Treatment recommendations for type and schedule of chemotherapeutic agents

Even if the decision is made to add chemotherapy to the treatment, the type and the schedule of chemotherapeutic agents must be determined. The goal is to determine the optimal timing and regimen, thereby maximizing the effectiveness of the treatment while minimizing the adverse effects. Numerous clinical trials to address this issue are ongoing.

Conventional versus altered fractionation, stereotactic radiation boost, and brachytherapy

The goal of these therapies is to find the optimal radiation regimen, thereby maximizing the effectiveness of this treatment while minimizing the adverse effects. The general recommendation for treatment of a primary tumor is a radiation dosage of at least 66 Gy. Stereotactic radiotherapy and brachytherapy may be used to boost dosage as well as to minimize surrounding tissue damage. Various clinical trials that involve different radiation regimens have been reported, and many more clinical trials are ongoing.

Salvage nasopharyngectomy or re-irradiation for local recurrence

The choice of therapy for local recurrence is another area of ongoing controversy. Fee concluded that the results of surgical resection are probably only slightly better than retreatment with radiotherapy. However, Fee believes that surgery is associated with fewer long-term complications when compared with re-irradiation. With the continued improvement in radiation delivery techniques such as intensity-modulated radiation therapy (IMRT) and stereotactic boost, complications associated with re-irradiation may decrease.

The best approach for performing nasopharyngectomy

None of the surgical approaches for resection of recurrent NPC is ideal. Because of the nature of the disease process, which involves an extremely complex anatomical region, the surgeon needs to be familiar with all of the surgical approaches. The operation must be tailored to the areas involved by the tumor and may involve a combination of approaches, thus allowing maximal exposure while minimizing associated morbidity.

Guidelines Summary

Guidelines for the treatment of nasopharyngeal carcinoma (NPC), issued by the American College of Radiology (ACR) in 2015, include the following recommendations[71] :

Author

Ho-Sheng Lin, MD, FACS, Professor and Interim Chair, Department of Otolaryngology-Head and Neck Surgery, Faculty, Sleep Fellowship Program, Divison of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Wayne State University School of Medicine; Chief, Section of Otolaryngology, Department of Surgery, John D Dingell Veterans Affairs Medical Center

Disclosure: Received consulting fee from Intuitive Surgical for proctoring; consultant for Checkpoint Surgical.

Coauthor(s)

Willard E Fee, Jr, MD, Edward C and Amy H Sewall Professor Emeritus, Chairman Emeritus, Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Nader Sadeghi, MD, FRCSC, Professor and Chairman, Department of Otolaryngology-Head and Neck Surgery, McGill University Faculty of Medicine; Chief Otolaryngologist, MUHC; Director, McGill Head and Neck Cancer Program, Royal Victoria Hospital, Canada

Disclosure: Nothing to disclose.

Chief Editor

Arlen D Meyers, MD, MBA, Professor of Otolaryngology, Dentistry, and Engineering, University of Colorado School of Medicine

Disclosure: Serve(d) as a director, officer, partner, employee, advisor, consultant or trustee for: Cerescan;RxRevu;Cliexa;The Physicians Edge;Sync-n-Scale;mCharts<br/>Received income in an amount equal to or greater than $250 from: The Physicians Edge, Cliexa<br/> Received stock from RxRevu; Received ownership interest from Cerescan for consulting; .

Additional Contributors

Benoit J Gosselin, MD, FRCSC, Associate Professor of Surgery, Dartmouth Medical School; Director, Comprehensive Head and Neck Oncology Program, Norris Cotton Cancer Center; Staff Otolaryngologist, Division of Otolaryngology-Head and Neck Surgery, Dartmouth-Hitchcock Medical Center

Disclosure: Nothing to disclose.

References

  1. Strazzulla A, Barreca GS, Giancotti A, et al. Nasopharyngeal carcinoma: review of the literature with a focus on therapeutical implications. Infez Med. 2015 Sep. 23 (3):224-9. [View Abstract]
  2. Fee WE Jr, Gilmer PA, Goffinet DR. Surgical management of recurrent nasopharyngeal carcinoma after radiation failure at the primary site. Laryngoscope. 1988 Nov. 98(11):1220-6. [View Abstract]
  3. Tu GY, Hu YH, Xu GZ, Ye M. Salvage surgery for nasopharyngeal carcinoma. Arch Otolaryngol Head Neck Surg. 1988 Mar. 114(3):328-9. [View Abstract]
  4. Hsu MM, Tu SM. Nasopharyngeal carcinoma in Taiwan. Clinical manifestations and results of therapy. Cancer. 1983 Jul 15. 52(2):362-8. [View Abstract]
  5. Chua DT, Sham JS, Choy D, et al. Patterns of failure after induction chemotherapy and radiotherapy for locoregionally advanced nasopharyngeal carcinoma: the Queen Mary Hospital experience. Int J Radiat Oncol Biol Phys. 2001 Apr 1. 49(5):1219-28. [View Abstract]
  6. Kwong DL, Sham JS, Au GK, et al. Concurrent and adjuvant chemotherapy for nasopharyngeal carcinoma: a factorial study. J Clin Oncol. 2004 Jul 1. 22(13):2643-53. [View Abstract]
  7. Leung TW, Tung SY, Sze WK, et al. Treatment results of 1070 patients with nasopharyngeal carcinoma: an analysis of survival and failure patterns. Head Neck. 2005 Jul. 27(7):555-65. [View Abstract]
  8. Perri F, Bosso D, Buonerba C, Lorenzo GD, Scarpati GD. Locally advanced nasopharyngeal carcinoma: Current and emerging treatment strategies. World J Clin Oncol. 2011 Dec 10. 2(12):377-83. [View Abstract]
  9. Tang LL, Chen WQ, Xue WQ, et al. Global trends in incidence and mortality of nasopharyngeal carcinoma. Cancer Lett. 2016 Jan 29. [View Abstract]
  10. Challapalli SD, Simpson MC, Adjei Boakye E, et al. Survival differences in nasopharyngeal carcinoma among racial and ethnic minority groups in the United States: a retrospective cohort study. Clin Otolaryngol. 2018 Sep 14. [View Abstract]
  11. Teo PM, Tsao SY, Ho JH, Yu P. A proposed modification of the Ho stage-classification for nasopharyngeal carcinoma. Radiother Oncol. 1991 May. 21(1):11-23. [View Abstract]
  12. Low WK, Leong JL, Goh YH, Fong KW. Diagnostic value of Epstein-Barr viral serology in nasopharyngeal carcinoma. Otolaryngol Head Neck Surg. 2000 Oct. 123(4):505-7. [View Abstract]
  13. Cai YL, Li J, Lu AY, Zheng YM, Zhong WM, Wang W, et al. Diagnostic significance of combined detection of Epstein-Barr virus antibodies, VCA/IgA, EA/IgA, Rta/IgG and EBNA1/IgA for nasopharyngeal carcinoma. Asian Pac J Cancer Prev. 2014. 15(5):2001-6. [View Abstract]
  14. Liu Z, Ji MF, Huang QH, Fang F, Liu Q, Jia WH, 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. 2013 Feb 1. 177(3):242-50. [View Abstract]
  15. Chan KC, Hung EC, Woo JK, Chan PK, Leung SF, Lai FP, et al. Early detection of nasopharyngeal carcinoma by plasma Epstein-Barr virus DNA analysis in a surveillance program. Cancer. 2013 May 15. 119(10):1838-44. [View Abstract]
  16. Ji MF, Huang QH, Yu X, Liu Z, Li X, Zhang LF, et al. Evaluation of plasma Epstein-Barr virus DNA load to distinguish nasopharyngeal carcinoma patients from healthy high-risk populations in Southern China. Cancer. 2014 May 1. 120(9):1353-60. [View Abstract]
  17. Yu E, O'Sullivan B, Kim J, Siu L, Bartlett E. Magnetic resonance imaging of nasopharyngeal carcinoma. Expert Rev Anticancer Ther. 2010 Mar. 10(3):365-75. [View Abstract]
  18. Ng SH, Chan SC, Yen TC, et al. Pretreatment evaluation of distant-site status in patients with nasopharyngeal carcinoma: accuracy of whole-body MRI at 3-Tesla and FDG-PET-CT. Eur Radiol. 2009 Jul 9. [View Abstract]
  19. Chua ML, Ong SC, Wee JT, et al. Comparison of 4 modalities for distant metastasis staging in endemic nasopharyngeal carcinoma. Head Neck. 2009 Mar. 31(3):346-54. [View Abstract]
  20. Liu T, Xu W, Yan WL, Ye M, Bai YR, Huang G. FDG-PET, CT, MRI for diagnosis of local residual or recurrent nasopharyngeal carcinoma, which one is the best? A systematic review. Radiother Oncol. 2007 Dec. 85(3):327-35. [View Abstract]
  21. Waxman AD. Use of thallium 201 in tumor evaluation. West J Med. 1992 Jul. 157(1):60. [View Abstract]
  22. Yen RF, Ting LL, Cheng MF, Wu YW, Tzen KY, Hong RL. Usefulness of 201TL SPECT/CT relative to 18F-FDG PET/CT in detecting recurrent skull base nasopharyngeal carcinoma. Head Neck. 2009 Jun. 31(6):717-24. [View Abstract]
  23. Shanmugaratnam K, Chan SH, de-The G, et al. Histopathology of nasopharyngeal carcinoma: correlations with epidemiology, survival rates and other biological characteristics. Cancer. 1979 Sep. 44(3):1029-44. [View Abstract]
  24. Fee WE Jr. Nasopharynx. Close LG, Larson DL, Shah JP. Essentials of Head and Neck Oncology. New York, NY: Thieme; 1998. 205-210.
  25. Chen CL, Wen WN, Chen JY, Hsu MM, Hsu HC. Detection of Epstein-Barr virus genome in nasopharyngeal carcinoma by in situ DNA hybridization. Intervirology. 1993. 36(2):91-8. [View Abstract]
  26. Hao SP, Tsang NM, Chang KP. Differentiation of recurrent nasopharyngeal carcinoma and skull base osteoradionecrosis by Epstein-Barr virus-derived latent membrane protein-1 gene. Laryngoscope. 2001 Apr. 111(4 Pt 1):650-2. [View Abstract]
  27. Kwong DL, Nicholls J, Wei WI, et al. The time course of histologic remission after treatment of patients with nasopharyngeal carcinoma. Cancer. 1999 Apr 1. 85(7):1446-53. [View Abstract]
  28. Tsai ST, Jin YT, Mann RB, Ambinder RF. Epstein-Barr virus detection in nasopharyngeal tissues of patients with suspected nasopharyngeal carcinoma. Cancer. 1998 Apr 15. 82(8):1449-53. [View Abstract]
  29. Dictor M, Siven M, Tennvall J, Rambech E. Determination of nonendemic nasopharyngeal carcinoma by in situ hybridization for Epstein-Barr virus EBER1 RNA: sensitivity and specificity in cervical node metastases. Laryngoscope. 1995 Apr. 105(4 Pt 1):407-12. [View Abstract]
  30. Al-Sarraf M, LeBlanc M, Giri PG, Fu KK, Cooper J, Vuong T. Chemoradiotherapy versus radiotherapy in patients with advanced nasopharyngeal cancer: phase III randomized Intergroup study 0099. J Clin Oncol. 1998 Apr. 16(4):1310-7. [View Abstract]
  31. Lu H, Peng L, Yuan X, et al. Concurrent chemoradiotherapy in locally advanced nasopharyngeal carcinoma: a treatment paradigm also applicable to patients in Southeast Asia. Cancer Treat Rev. 2009 Jun. 35(4):345-53. [View Abstract]
  32. Al-Sarraf M. Treatment of locally advanced head and neck cancer: historical and critical review. Cancer Control. 2002 Sep-Oct. 9(5):387-99. [View Abstract]
  33. Chi KH, Chang YC, Guo WY, et al. A phase III study of adjuvant chemotherapy in advanced nasopharyngeal carcinoma patients. Int J Radiat Oncol Biol Phys. 2002 Apr 1. 52(5):1238-44. [View Abstract]
  34. Tam M, Lee A, Wu SP, et al. Neoadjuvant chemotherapy in local-regionally advanced nasopharyngeal carcinoma: A National Cancer Database analysis. Laryngoscope. 2018 Aug 22. [View Abstract]
  35. Chua DT, Sham JS, Choy D, et al. Preliminary report of the Asian-Oceanian Clinical Oncology Association randomized trial comparing cisplatin and epirubicin followed by radiotherapy versus radiotherapy alone in the treatment of patients with locoregionally advanced nasopharyngeal carcinoma. Asian-Oceanian Clinical Oncology Association Nasopharynx Cancer Study Group. Cancer. 1998 Dec 1. 83(11):2270-83. [View Abstract]
  36. Ma BB, Chan AT. Recent perspectives in the role of chemotherapy in the management of advanced nasopharyngeal carcinoma. Cancer. 2005 Jan 1. 103(1):22-31. [View Abstract]
  37. Chan AT, Leung SF, Ngan RK, et al. Overall survival after concurrent cisplatin-radiotherapy compared with radiotherapy alone in locoregionally advanced nasopharyngeal carcinoma. J Natl Cancer Inst. 2005 Apr 6. 97(7):536-9. [View Abstract]
  38. Lin JC, Jan JS, Hsu CY, Liang WM, Jiang RS, Wang WY. Phase III study of concurrent chemoradiotherapy versus radiotherapy alone for advanced nasopharyngeal carcinoma: positive effect on overall and progression-free survival. J Clin Oncol. 2003 Feb 15. 21(4):631-7. [View Abstract]
  39. Wee J, Tan EH, Tai BC, 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 Sep 20. 23(27):6730-8. [View Abstract]
  40. Wee J. 4th FY Khoo Memorial Lecture 2008: Nasopharyngeal Cancer Workgroup--the past, the present and the future. Ann Acad Med Singapore. 2008 Jul. 37(7):606-14. [View Abstract]
  41. Lee AW, Lau WH, Tung SY, Chua DT, Chappell R, Xu L. Preliminary results of a randomized study on therapeutic gain by concurrent chemotherapy for regionally-advanced nasopharyngeal carcinoma: NPC-9901 Trial by the Hong Kong Nasopharyngeal Cancer Study Group. J Clin Oncol. 2005 Oct 1. 23(28):6966-75. [View Abstract]
  42. Lee AW, Tung SY, Chua DT, Ngan RK, Chappell R, Tung R, et al. Randomized trial of radiotherapy plus concurrent-adjuvant chemotherapy vs radiotherapy alone for regionally advanced nasopharyngeal carcinoma. J Natl Cancer Inst. 2010 Aug 4. 102(15):1188-98. [View Abstract]
  43. Chen Y, Liu MZ, Liang SB, et al. Preliminary results of a prospective randomized trial comparing concurrent chemoradiotherapy plus adjuvant chemotherapy with radiotherapy alone in patients with locoregionally advanced nasopharyngeal carcinoma in endemic regions of china. Int J Radiat Oncol Biol Phys. 2008 Aug 1. 71(5):1356-64. [View Abstract]
  44. Chen Y, Sun Y, Liang SB, Zong JF, Li WF, 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 Jun 15. 119(12):2230-8. [View Abstract]
  45. Huncharek M, Kupelnick B. Combined chemoradiation versus radiation therapy alone in locally advanced nasopharyngeal carcinoma: results of a meta-analysis of 1,528 patients from six randomized trials. Am J Clin Oncol. 2002 Jun. 25(3):219-23. [View Abstract]
  46. Baujat B. Chemotherapy as an adjunct to radiotherapy in locally advanced nasopharyngeal carcinoma. Cochrane Database Syst Rev. Oct 18, 2006. 4:[View Abstract]
  47. Chen L, Hu CS, Chen XZ, Hu GQ, Cheng ZB, 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 Feb. 13(2):163-71. [View Abstract]
  48. Liang ZG, Zhu XD, Zhou ZR, Qu S, Du YQ, Jiang YM. Comparison of concurrent chemoradiotherapy followed by adjuvant chemotherapy versus concurrent chemoradiotherapy alone in locoregionally advanced nasopharyngeal carcinoma: a meta-analysis of 793 patients from 5 randomized controlled trials. Asian Pac J Cancer Prev. 2012. 13(11):5747-52. [View Abstract]
  49. Kong L, Hu C, Niu X, Zhang Y, Guo Y, Tham IW, et al. Neoadjuvant chemotherapy followed by concurrent chemoradiation for locoregionally advanced nasopharyngeal carcinoma: interim results from 2 prospective phase 2 clinical trials. Cancer. 2013 Dec 1. 119(23):4111-8. [View Abstract]
  50. Preliminary results of a randomized trial comparing neoadjuvant chemotherapy (cisplatin, epirubicin, bleomycin) plus radiotherapy vs. radiotherapy alone in stage IV(> or = N2, M0) undifferentiated nasopharyngeal carcinoma: a positive effect on progression-free survival. International Nasopharynx Cancer Study Group. VUMCA I trial. Int J Radiat Oncol Biol Phys. 1996 Jun 1. 35(3):463-9. [View Abstract]
  51. Hareyama M, Sakata K, Shirato H, et al. A prospective, randomized trial comparing neoadjuvant chemotherapy with radiotherapy alone in patients with advanced nasopharyngeal carcinoma. Cancer. 2002 Apr 15. 94(8):2217-23. [View Abstract]
  52. Rossi A, Molinari R, Boracchi P, et al. Adjuvant chemotherapy with vincristine, cyclophosphamide, and doxorubicin after radiotherapy in local-regional nasopharyngeal cancer: results of a 4-year multicenter randomized study. J Clin Oncol. 1988 Sep. 6(9):1401-10. [View Abstract]
  53. Al-Sarraf M, LeBlanc M, Giri PG. Superiority of 5-year survival with chemoradiotherapy vs. radiotherapy in patients with locally advanced nasopharyngeal cancer. Intergroup 0099 phase III study: final report abstract. Proc Am Soc Clin Oncol. 2001. 20:227a.
  54. Lee AW, Poon YF, Foo W, et al. Retrospective analysis of 5037 patients with nasopharyngeal carcinoma treated during 1976-1985: overall survival and patterns of failure. Int J Radiat Oncol Biol Phys. 1992. 23(2):261-70. [View Abstract]
  55. Sham JS, Choy D. Prognostic factors of nasopharyngeal carcinoma: a review of 759 patients. Br J Radiol. 1990 Jan. 63(745):51-8. [View Abstract]
  56. Fu KK, Newman H, Phillips TL. Treatment of locally recurrent carcinoma of the nasopharynx. Radiology. 1975 Nov. 117(2):425-31. [View Abstract]
  57. Yan JH, Hu YH, Gu XZ. Radiation therapy of recurrent nasopharyngeal carcinoma. Report on 219 patients. Acta Radiol Oncol. 1983. 22(1):23-8. [View Abstract]
  58. Zhang EP, Lian PG, Cai KL, et al. Radiation therapy of nasopharyngeal carcinoma: prognostic factors based on a 10-year follow-up of 1302 patients. Int J Radiat Oncol Biol Phys. 1989 Feb. 16(2):301-5. [View Abstract]
  59. Wei WI, Ho CM, Wong MP, Ng WF, Lau SK, Lam KH. Pathological basis of surgery in the management of postradiotherapy cervical metastasis in nasopharyngeal carcinoma. Arch Otolaryngol Head Neck Surg. 1992 Sep. 118(9):923-9; discussion 930. [View Abstract]
  60. Na'ara S, Amit M, Billan S, Cohen JT, Gil Z. Outcome of Patients Undergoing Salvage Surgery for Recurrent Nasopharyngeal Carcinoma: A Meta-analysis. Ann Surg Oncol. 2014 Apr 18. [View Abstract]
  61. Fisch U. The infratemporal fossa approach for nasopharyngeal tumors. Laryngoscope. 1983 Jan. 93(1):36-44. [View Abstract]
  62. Panje WR, Gross CE. Treatment of tumor of the nasopharynx: surgical therapy. Thawley SE, Panje WR. Comprehensive Management of Head and Neck Tumors. Philadelphia, Pa: WH Sauders Co; 1987. 1: 662-683.
  63. Wei WI, Ho CM, Yuen PW, Fung CF, Sham JS, Lam KH. Maxillary swing approach for resection of tumors in and around the nasopharynx. Arch Otolaryngol Head Neck Surg. 1995 Jun. 121(6):638-42. [View Abstract]
  64. Morton RP, Liavaag PG, McLean M, Freeman JL. Transcervico-mandibulo-palatal approach for surgical salvage of recurrent nasopharyngeal cancer. Head Neck. 1996 Jul-Aug. 18(4):352-8. [View Abstract]
  65. King WW, Ku PK, Mok CO, Teo PM. Nasopharyngectomy in the treatment of recurrent nasopharyngeal carcinoma: a twelve-year experience. Head Neck. 2000 May. 22(3):215-22. [View Abstract]
  66. Tsang RK, To VS, Ho AC, Ho WK, Chan JY, Wei WI. Early results of robotic assisted nasopharyngectomy for recurrent nasopharyngeal carcinoma. Head Neck. 2014 Mar 7. [View Abstract]
  67. Young YH. Irradiated ears in nasopharyngeal carcinoma survivors: a review. Laryngoscope. 2018 Sep 8. [View Abstract]
  68. Cheng SW, Ting AC, Lam LK, Wei WI. Carotid stenosis after radiotherapy for nasopharyngeal carcinoma. Arch Otolaryngol Head Neck Surg. 2000 Apr. 126(4):517-21. [View Abstract]
  69. McDowell LJ, Rock K, Xu W, et al. Long-Term Late Toxicity, Quality of Life, and Emotional Distress in Patients With Nasopharyngeal Carcinoma Treated With Intensity Modulated Radiation Therapy. Int J Radiat Oncol Biol Phys. 2018 Oct 1. 102 (2):340-52. [View Abstract]
  70. Liao D, Huang H, Zhu Z, et al. Prognostic value of matrix metalloproteinase 9 in nasopharyngeal carcinoma: a meta-analysis. Minerva Med. 2016 Jan 22. [View Abstract]
  71. [Guideline] Saba NF, Salama JK, Beitler JJ, et al. Nasopharyngeal carcinoma. American College of Radiology. Available at https://acsearch.acr.org/docs/3094110/Narrative/. 2015; Accessed: Oct 7, 2016.
  72. Co JL, Mejia MBA, Dizon JMR. Evidence on Effectiveness of Upper Neck Irradiation Versus Whole Neck Irradiation as Elective Neck Irradiation in Node-Negative Nasopharyngeal Cancer: A Meta-Analysis. J Glob Oncol. 2018 Sep. 1-11. [View Abstract]
  73. Lydiatt W, O'Sullivan B, Patel S. Major Changes in Head and Neck Staging for 2018. Am Soc Clin Oncol Educ Book. 2018 May 23. 505-14. [View Abstract]

Axial T2-weighted image shows a left-sided cervical nodal metastasis resulting from nasopharyngeal cancer.

Axial T2-weighted image shows a left-sided cervical nodal metastasis resulting from nasopharyngeal cancer.

Serology Status Sensitivity Specificity PPV NPV
IgA to EA80.2%100%100%83.5%
IgA to VCA97.3%46.8%64.7%94.5%
IgA Antibody to EA IgA Antibody to VCA Probability of NPC
++100%
+100%
5.5%
+37.8%
Serology Status Sensitivity Specificity Area Under ROC Curve (95% CI)
IgA to EA89.1%98.5%0.94 (0.92-0.96) 
IgA to VCA98.1%82.8%0.98 (0.96-0.99)  
IgG to Rta90.5%85.2%0.92 (0.89-0.95)  
IgA to EBNA187.2%84.2%0.92 (0.89-0.95)  
Combination Sensitivity Specificity Area Under ROC Curve (95% CI)
IgG to Rta +



IgA to EBNA1



93.4%90.6%0.97 (0.95-0.98) 
IgA to VCA +



IgA to EA



92.4%98.5%0.98 (0.96-0.99)  
IgA to VCA +



IgA to EBNA1



94.3%98.0%0.98 (0.97-0.99)  
IgA to VCA +



IgG to Rta



94.8%98.0%0.99 (0.98-1.00)  
IgA to VCA +



IgA to EA +



IgA to EBNA1



97.2%95.6%0.98 (0.97-0.99)  
IgA to VCA +



IgA to EA +



IgG to Rta



92.9%99.5%0.99 (0.98-1.00)  
IgA to VCA +



IgG to Rta +



IgA to EBNA1



94.8%98.5%0.99 (0.98-1.00)  
IgA to VCA +



IgA to EA +



IgG to Rta +



IgA to EBNA1



96.7%97.0%0.99 (0.98-1.00)  
 



Author, Year



Stage Number



of



Patients



Treatment Arms Survival Rate P Value
Neoadjuvant Chemotherapy Followed by Radiation
VUMCA, 1996[50] IVn=171Bleomycin/epirubicin/cisplatin X 3



Radiation



60% (3 yr OS)P > .05
n=168Radiation alone54% (3 y OS)
Hareyama, 2002[51] I-IVn=40Cisplatin/5-FU X 2



Radiation



60% (5 y OS)P > .05
n=40Radiation alone45% (5 y OS)
Chua, 1998[35] T3



N2-3



n=167Cisplatin/epirubicin X 2-3



Radiation



78% (3 y OS)P = .57
n=167Radiation alone71% (3 y OS)
Ma, 2001[36] III-IVn=224Cisplatin/bleomycin/5-FU X 2-3



Radiation



63% (5 y OS)P = .11
n=225Radiation alone56% (5 y OS)
Concurrent Chemotherapy and Radiation
Lin, 2003[38] III-IVn=141Cisplatin/5-FU X 2 +



Radiation



72.3% (5 y OS)P = .002
n=143Radiation alone54.2% (5 y OS)
Chan, 2005[37] II-IVn=174Cisplatin weekly and



Radiation



70.3% (5 y OS)P = .048
n=176Radiation alone58.6% (5 y OS)
Radiation Followed by Adjuvant Chemotherapy
Rossi, 1988[52] I-IVn=113Radiation



Vincristine/cyclophosphamide/Adriamycin X 6



59% (4 y OS)P > .05
n=116Radiation alone67% (4 y OS)
Chi, 2002[33] IVn=77Radiation



Cisplatin/5-FU/leucovorin X 9



61% (5 y OS)P = .5
n=77Radiation alone55% (5 y OS)
Neoadjuvant Chemotherapy Followed by Radiation Followed by Adjuvant Chemotherapy
Chan, 1995[37]  n=34Cisplatin/5-FU X 2



Radiation



Cisplatin/5-FU X 4



80% (5 y OS)P = .1
n=40Radiation alone81% (5 y OS)
Concurrent Chemotherapy and Radiation Followed by Adjuvant Chemotherapy
Al-Sarraf, 1998[30] III-IVn=93Cisplatin X 3 +



Radiation



Cisplatin/5-FU X 3



78% (3 y OS)P< 0.001
n=92Radiation alone47% (3 y OS)
Al-Sarraf, 2002[32] ; 2001[53] III-IVn=93Cisplatin X 3 +



Radiation



Cisplatin/5-FU X 3



67% (5 y OS)P< 0.001
n=92Radiation alone37% (5 y OS)
Wee, 2004[39, 40] III-IVn=111Cisplatin X 3 +



Radiation



Cisplatin/5-FU X 3



67% (5 y OS)P = 0.008
n=110Radiation alone49% (5 y OS)
Lee, 2005[41] ; 2010[42] T1-4



N2-3



n=172Cisplatin X 3 +



Radiation



Cisplatin/5-FU X 3



68% (5 y OS)P = 0.22
n=176Radiation alone64% (5 y OS)
Chen, 2013[44] III-IVn=1581) Cisplatin weekly + radiation



2) Cisplatin/5-FU X 3



72% (5 y OS)P = 0.043
n=158Radiation alone
62% (5 y OS)
Treatment WHO I, II, III



(n=147) OS, %



WHO II and III



(n=111, 75) OS, %



WHO I



(n=36, 25%) OS, %



Radiation374514
Chemoradiation677059