You are on page 1of 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/281635294

Management of Nasopharyngeal Carcinoma: Current Practice and Future


Perspective

Article  in  Journal of Clinical Oncology · September 2015


DOI: 10.1200/JCO.2015.60.9347

CITATIONS READS

122 438

4 authors, including:

Anne W M Lee Brigette B Y Ma


The University of Hong Kong The Chinese University of Hong Kong
216 PUBLICATIONS   6,625 CITATIONS    232 PUBLICATIONS   6,450 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Nasopharyngeal carcinoma View project

All content following this page was uploaded by Anne W M Lee on 15 August 2018.

The user has requested enhancement of the downloaded file.


VOLUME 33 䡠 NUMBER 29 䡠 OCTOBER 10 2015

JOURNAL OF CLINICAL ONCOLOGY R E V I E W A R T I C L E

Management of Nasopharyngeal Carcinoma: Current


Practice and Future Perspective
Anne W.M. Lee, Brigette B.Y. Ma, Wai Tong Ng, and Anthony T.C. Chan
Anne W.M. Lee, University of Hong
Kong–Shenzhen Hospital, Shenzhen; A B S T R A C T
Brigette B.Y. Ma and Anthony T.C.
Chan, Chinese University of Hong Nasopharyngeal carcinoma of the undifferentiated subtype is endemic to southern China, and
Kong; and Wai Tong Ng, Pamela Youde patient prognosis has improved significantly over the past three decades because of advances
Nethersole Eastern Hospital, Hong in disease management, diagnostic imaging, radiotherapy technology, and broader application
Kong Special Administrative Region,
of systemic therapy. Despite the excellent local control with modern radiotherapy, distant
People’s Republic of China.
failure remains a key challenge. Advances in molecular technology have helped to decipher the
Published online ahead of print at molecular pathogenesis of nasopharyngeal carcinoma as well as its etiologic association with
www.jco.org on September 8, 2015.
the Epstein-Barr virus. This in turn has led to the discovery of novel biomarkers and drug
Authors’ disclosures of potential targets, rendering this cancer site a current focus for new drug development. This article
conflicts of interest are found in the
reviews and appraises the key literature on the current management of nasopharyngeal
article online at www.jco.org. Author
contributions are found at the end of
carcinoma and future directions in clinical research.
this article.
J Clin Oncol 33:3356-3364. © 2015 by American Society of Clinical Oncology
Corresponding author: Anthony T.C.
Chan, MD, State Key Laboratory in
Oncology in South China, Sir Y.K. Pao with advances in diagnostic radiology and treat-
Centre for Cancer, Department of Clini- INTRODUCTION
ment. There are three areas in the current (seventh)
cal Oncology, Hong Kong Cancer Insti-
tute, Chinese University of Hong Kong, Nasopharyngeal carcinoma (NPC) of the undiffer- edition of the AJCC/UICC staging criteria for NPC
Hong Kong SAR, China; e-mail: entiated subtype remains endemic in southern that may warrant revision in the forthcoming
anthony@clo.cuhk.edu.hk. (eighth) edition. First, the current criterion of N3b
China, with a peak annual incidence approaching 30
© 2015 by American Society of Clinical per 100,000 persons. Overall prognosis has dramat- stage is defined as disease extension to the supracla-
Oncology
ically improved over the past three decades because vicular fossa—a triangular region defined by the
0732-183X/15/3329w-3356w/$20.00 superior margin of the sternal end of the clavicle, the
of advances in management, including the improve-
DOI: 10.1200/JCO.2015.60.9347 ment of radiotherapy (RT) technology, the broader superior margin of the lateral end of the clavicle, and
application of chemotherapy, and more accurate the point where the neck meets the shoulder. How-
disease staging. Current therapeutic decisions are ever, this definition, which was originally proposed
based mainly on disease stage.1,2 The general con- by Ho,3 is based primarily on clinical examination.
sensus is to treat stage I disease with RT alone, stage There are now substantial data to support that this
II disease with RT with or without concurrent che- region can be extended from the supraclavicular
motherapy (CRT), and stage III to IVB NPC with fossa to lower levels in the neck, including levels IV
CRT. Excellent local control can generally be and Vb. This broader definition enables a more re-
achieved, but distant failure is a key problem, and liable radiologic assessment without affecting the
the treatment outcome of metastatic NPC remains overall prognostic significance of the N3b stage.4,5
disappointing. Another challenge is achieving suc- The second area concerns the definition of T4 dis-
cessful local control with minimal late toxicity, espe- ease as being an invasion of the masticator space,
cially for tumors with intracranial extension. This which was originally intended as a synonym for the
article reviews current challenges and future per- infratemporal fossa, as stated in the sixth edition of
spectives in managing this unique cancer. the AJCC/UICC staging criteria.6 Clarification and
redefinition of this criterion are needed, because pa-
tients with tumor extension confined to the ptery-
STAGING AND PROGNOSTIC FACTORS goid muscles generally have a prognosis that is more
comparable to that of patients with T2 disease rather
Despite its known limitations, TNM staging remains those with T4 disease.7 The third area is that the
the most important prognostic factor in NPC. Con- prognostic differentiation between stage T1 and T3
tinual revision of the American Joint Committee on disease is narrowing with modern treatment; there-
Cancer (AJCC)/International Union Against Can- fore, regrouping is needed to better reflect the results
cer (UICC) staging system is needed to keep pace of contemporary practice.8

3356 © 2015 by American Society of Clinical Oncology

Downloaded from ascopubs.org by University OF HONG KONG Libraries on August 14, 2018 from 147.008.031.043
Copyright © 2018 American Society of Clinical Oncology. All rights reserved.
Management of Nasopharyngeal Carcinoma

Besides using anatomic end points in the staging of NPC, there is To minimize the risk of geographic miss resulting from the highly
increasing evidence that primary tumor volume could be a significant conformal dose distribution of IMRT, accuracy in the delineation of
factor for local control independent of T stage,8 and lactate dehydro- gross tumor volume (GTV), appropriate design of margins and clini-
genase could be a significant factor for distant control independent of cal target volume, and precision in RT delivery are important stan-
stage group.9 Among the molecular markers, the most studied is dards to maintain when prescribing IMRT. Thorough pretreatment
plasma Epstein-Barr virus (EBV) DNA, which is universally associated assessment incorporating endoscopy, magnetic resonance imaging,
with the nonkeratinizing subtype of NPC. In addition to being a good and positron emission tomography coupled with computed tomog-
prognosticator, it is also useful for assessing treatment response and raphy (CT), as well as fusion of images from different modalities with
detecting disease relapse.10-12 Novel prognostic markers using genetic the planning CT images, is fundamental. International protocols, such
signatures and microRNA have also shown promising results.13,14 as those from the Radiation Therapy Oncology Group,27 represent
useful and practical references, and regular evaluation of treatment
ADVANCES IN RT outcome and patterns of failure should be conducted in centers that
practice IMRT to optimize the planning parameters.
The development of image-guided RT from conventional portal
Intensity-modulated RT (IMRT) is the current standard of treatment
films may improve treatment precision by allowing the checking of
for NPC. With conformity of dose distribution achieved by sculpting
the high-dose zone for complete coverage of tumor targets while bony structures through onboard images, the cross checking of both
sparing the critical normal structures, excellent locoregional control bone and soft tissues by cone-beam CT, and real-time infra-fraction
can be achieved by most expert centers15-20,22 (Table 1). Random- monitoring. A substantial shrinkage of tumor and parotid glands as
ized controlled trials comparing IMRT with conventional two- well as changes in body contour may occur during the course of RT,
dimensional RT have shown a significant reduction in the risk of leading to shifts in dose distribution to both the tumor target and
permanent xerostomia, especially in patients with early disease,23,24 as normal structures.28-30 Adaptive RT consisting of reassessment and
well as improvement in local control with reduction of other treat- replanning (when indicated) during the course of RT is hence increas-
ment complications.16,21 One major refinement of the delineation of ingly advocated to ensure the continual appropriateness of dose dis-
high-risk regions for elective irradiation is the tailoring of clinical tribution. Improvements in locoregional control and quality of life
tumor volume for node-negative patients. In the past, elective RT was have been observed with this approach,31 but the optimal timing for
recommended for the bilateral whole neck. There is increasing evi- treatment replanning remains an open question. Although technolo-
dence that the lymphatic spread of NPC follows a fairly predictable gies such as in–treatment room imaging, deformable image registra-
order, with retropharyngeal nodes and the level II neck nodes being tion, and knowledge-based treatment planning tools are being used to
most commonly affected and skip metastases being uncommon. Ret- enhance the efficiency of adaptive RT, it is still a labor-intensive pro-
rospective and randomized studies have shown that selective neck cess. Furthermore, cost effectiveness and long-term impact on thera-
irradiation confined to levels II, III, and Va is safe and would not peutic ratios remain to be determined.
jeopardize nodal control or survival.25,26 This sparing of the lower Although IMRT can achieve excellent 5-year local control rates
neck and level IB is useful, because the latter allows a better sparing of ⱖ 90% for T3 disease, the corresponding rates for T4 disease still range
the submandibular gland, thus reducing the risk of xerostomia. from 74% to 80% (Table 1).16,18-20,22 Late toxicities, especially

Table 1. Five-Year Treatment Outcome With Intensity-Modulated Radiotherapy

Dose (Gy) T4 Disease (%)


Local Nodal Distant
No. of Per Chemotherapy Control Control Control OS Local Serious Neurologic
Study Patients Total Fraction (%) (%) (%) (%) (%) Patients Control Complications (%)
Lai et al15 512 NR 2.27 81 93 97 84 NR 52 (T3 to T4) 82 NR
Peng et al16 306 70 2.12 60 91 92 NR 80 17ⴱ 82 TLN, 13.1
CN, 3.9
OP, 1.6
Lin et al17 414 66 to 70.95 — 81 95 97 82 80 21 NR NR
30 to 33 fractions
Wu et al18 249 68 to 72 (30 to 32 fractions) — 100 87 88 78 78 33 85 (T3 to T4) TLN, 2.6
30 to 32 fractions CN, 1.5
OP, 1.8
Sun et al19 868 68 2.27 83 92 96 85 NR 19 83 TLN or BS, 5.5
Ng et al20 444 70 2 to 2.12 83 86 92 83 80 23 74 TLN, 0.5
CN, 1.621
Yi et al22 271 70 (T1 to T2) 2.12 52 87 NR 70 79 25 54 and 76† NR
74 (T3 to T4) 2.24

Abbreviations: BS, brainstem injury; CN, cranial nerve injury; NR, not reported; OP, optice nerve or chiasm injury; OS, overall survival; TLN, temporal lobe necrosis.

Randomized study; percentage is based on both treatment arms of 616 patients.
†With and without concurrent chemotherapy, respectively.

www.jco.org © 2015 by American Society of Clinical Oncology 3357

Downloaded from ascopubs.org by University OF HONG KONG Libraries on August 14, 2018 from 147.008.031.043
Copyright © 2018 American Society of Clinical Oncology. All rights reserved.
Lee et al

neurologic damage, are also more common after RT for T3 to T4 questions remain. These include the value of adding targeted ther-
NPC.16,19 The general principle is to administer the maximum tumor apy, induction, or adjuvant chemotherapy to CRT in different
dose within the tolerance of critical organs at risk. To optimize the sequences, the value of CRT in patients with stage II NPC, optimi-
balance between local control and complications, appropriate design zation of patient selection for CRT using predictive biomarkers,
of treatment dose constraints and more accurate data on the and evaluation of the long-term efficacy and safety of using con-
maximum-tolerated dose regarding organs at risk are needed. The current chemotherapy with IMRT.
target volume underdosed (⬍ 65 Gy) is one of the most significant The merit of adding induction chemotherapy to CRT in stage III
factors affecting tumor control and overall survival (OS).20 Hence, to IVB NPC is a subject of intense investigation. The study by Hui et
modifications of dose constraints with inclusion of the GTV as one of al50 in 2009 was the first to report an OS benefit with this approach.
the top priority parameters and acceptance of higher doses to less Although this study was not powered to detect a survival difference
critical structures (eg, temporal lobes and unilateral optic nerve) are between the study arms, the significant finding of a 26.5% absolute
now being tested cautiously. improvement in 3-year OS by adding induction cisplatin and do-
For large primary tumors abutting or infiltrating critical struc- cetaxel to CRT did provide a strong impetus for phase III validation.
tures, one common practice is to add induction chemotherapy to To date, at least six randomized studies have been initiated or reported
shrink the tumor bulk for better dose coverage during subsequent RT in centers in Hong Kong (ClinicalTrials.gov identifier NCT00379262),
plus concurrent cisplatin.32 Other novel strategies that warrant further Singapore (NCT00997906),51 Taiwan (NCT00201396), Europe
investigation include the use of more conformal techniques, such as (NCT00828386; terminated because of low accrual; also randomized
proton or heavy particle therapy,33 and the use of selective radio- phase II study52), and China (NCT01245959, NCT01536223). The stud-
sensitizing agents targeting the tumor and radio protectants targeting ies by Tan et al51 and Fountzilas et al,52 which investigated the use of
the organs at risk. The current practice is to prescribe a fairly uniform induction chemotherapy using a triplet regimen before CRT, failed to
and standardized dose level (approximately 70 Gy) to the whole GTV. achieve an OS benefit at 3 years. A six-arm study from the Hong Kong
Another future perspective is to individualize the radiation dose. With NPC study group (NCT00379262) was designed to address questions
the advent of functional positron emission tomography and magnetic concerning drug sequences, RT fractionation, and substitution of
resonance imaging, it may be possible to determine and deliver the
fluorouracil (FU) with capecitabine.53 A total of 803 patients with
most appropriate dose levels to different parts of the target volume
stage III to IVB NPC were randomly assigned. Using the Intergroup-
with IMRT, optimizing the therapeutic ratio for individual patients.
0099 regimen of concurrent cisplatin plus adjuvant cisplatin and FU as
the standard arm, preliminary analyses failed to meet the primary end
CRT FOR LOCOREGIONALLY ADVANCED DISEASE points of the study. Subgroup analyses suggested that oral capecitabine
may be a safe alternative to FU, and accelerated RT was not recom-
mended for patients undergoing CRT for locoregionally advanced
With almost half of all patients presenting at an advanced stage, of
NPC.53 It should be noted that the median follow-up durations of the
whom one third subsequently die as a result of this cancer within 5
reported studies are only approximately 3 years; thus, longer
years of diagnosis, the treatment of stage III to IVB NPC has
follow-up is needed before definitive conclusions can be drawn re-
deservedly been the main focus of clinical research in the past
decades.34 The improvement of clinical outcome with the use of garding the utility of adding induction chemotherapy to CRT. One
CRT is an important milestone in this endeavor. To date, at least could argue that the magnitude of any theoretic benefit would be
nine randomized studies have compared concurrent CRT (⫾ ad- small; this implies that a much larger phase III study is needed. An
juvant chemotherapy) versus RT alone.35-42 These studies have alternate explanation is that perhaps only specific subgroups would
reported a significant improvement in event-free survival, and in benefit from this approach, such as patients with early metabolic
some studies, an advantage in OS has been observed, with hazard response to induction chemotherapy.54 The cumulative evidence
ratios (HRs; where reported) for death ranging from 0.51 to 0.71 at on induction chemotherapy in stage III to IVB disease remains
3 to 5 years of follow-up35-37,39 (Table 2). This benefit has been insufficient to change practice at this juncture, and the results of
observed irrespective of the diverse types of chemotherapeutic other phase III studies are eagerly awaited.
regimens used in these studies. However, in some exploratory Among the recent highlights in research, the role of adjuvant
analyses, the dose of cisplatin delivered during the concurrent chemotherapy in stage III to IVB NPC and the use of CRT in stage II
phase has been shown to influence locoregional control and NPC warrant discussion. To date, no randomized phase III studies
OS.40,48 Subgroup analyses of some randomized studies have also have demonstrated a survival advantage with adjuvant chemotherapy.
suggested that the OS benefit may be larger in patients with T3 to In the meta-analysis by Blanchard et al,49 the magnitude of the OS
T4 disease.35,36 benefit observed in the subgroup receiving CRT plus adjuvant chemo-
In a recently presented meta-analysis, in which patient-based therapy (n ⫽ 1,267 patients; HR, 0.65; 95% CI, 0.56 to 0.76) seemed to
data on 4,798 patients with mostly stage III to IVB NPC were analyzed, be larger than that in the CRT-alone subgroup (n ⫽ 1,834 patients;
the addition of chemotherapy was found to confer an absolute OS HR, 0.80; 95% CI, 0.70 to 0.93). In the largest phase III study reported
benefit of 6% at 5 years and 8% at 10 years.49 The benefit was signifi- to date, Chen et al55 found that the addition of adjuvant cisplatin plus
cant for OS (HR, 0.79; 95% CI, 0.72 to 0.86), progression-free survival FU did not improve failure-free survival after a relatively short
(HR, 0.76; 95% CI, 0.70 to 0.82), locoregional control (HR, 0.74; 95% follow-up period of 2 years, with an HR of 0.74 (95% CI, 0.49 to 1.10;
CI, 0.65 to 0.85), and distant control (HR, 0.68; 95% CI, 0.60 to P ⫽ .13). Furthermore, compliance was a problem; only 63% of
0.76). Despite the widespread acceptance of CRT as the new ther- patients could complete the planned chemotherapy.55 Data from the
apeutic benchmark for stage III to IVB NPC, several unanswered current literature do not allow a definitive recommendation to be

3358 © 2015 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

Downloaded from ascopubs.org by University OF HONG KONG Libraries on August 14, 2018 from 147.008.031.043
Copyright © 2018 American Society of Clinical Oncology. All rights reserved.
www.jco.org
Table 2. Summary of Selected Phase III Studies Comparing Concurrent CRT Versus RT Alone

Study Arms
TNM Stage % Results
No. of Patients/ CRT RT Alone
Author Year II III IV Sample Size Treatment Dose (Gy) CRT (%) RT (%) HRⴱ 95% CI P
Al-Sarraf et al38 1998, 2001 9 91 147 Cisplatin every 3 weeks plus 70
5-year OS RT followed by cisplatin 67 37 .001
plus FU (n ⫽ 14)
5-year PFS 58 29 .001
Chan et al35,43 2002, 2005 28.8 29.4 41.8 350 Cisplatin once per week 66
5-year OS plus RT 72 59 0.71 0.5 to 1.0 .048
5-year PFS 62 52 0.74 0.54 to 1.0 .076
Lin et al44 2003 19.7 80.3 284 Cisplatin every 4 weeks plus 70 to 74
5-year OS FU and RT 72.3 54.2 .002
5-year PFS 71.6 53.0 .001
5-year DMFS 78.7 69.9 .057
Wee et al37 2005 1 45 54 221 Cisplatin every 3 weeks plus 70
3-year OS RT followed by cisplatin 80 65 0.51 0.31 to 0.81 .0061
plus FU
3-year DFS 72 53 0.57 0.38 to 0.87 .0093
2-year 30 13 .0029
relapse
rate
Lee et al45 2011 — 189 Cisplatin every 3 weeks plus 70†‡
3-year OS RT followed by cisplatin 87† and 88‡ 83† and 73‡ 0.52§ 0.28 to 0.97 NS
plus FU
3-year PFS 68† and 63‡ 73† and 88‡
Lee et al41,46 2010 59 41 348 Cisplatin every 3 weeks plus 70
5-year OS RT followed by cisplatin 68 64 0.81 0.58 to 1.13 .22
plus FU
5-year PFS 62 53 0.72 0.53 to 0.98 .035
5-year DFFR 74 68 .32
5-year LRFFR 88 78 .005
Chen et al42 2011 87 13 230 Cisplatin 30 mg/m2 once per 60 to 70
Management of Nasopharyngeal Carcinoma

5-year OS week 94.5 85.6 0.30 0.12 to 0.76 < .001


5-year DMFS 94.8 83.9 < .001
5-year LRS 93 91.1 .29
Zhang et al47 2005, 2013 115 Oxaliplatin plus RT 70 to 74
5-year OS 73.2 60.2 .03

Copyright © 2018 American Society of Clinical Oncology. All rights reserved.


5-year DMFS 74.3 63 .03
Chen et al39 2013 36 64 316 Cisplatin once per week 66
5-year OS plus RT followed by 72 62 0.69 0.48 to 0.99 .043
cisplatin plus FU
5-year PFS 68 57 .015

NOTE. Bold font indicates significance.


Abbreviations: CRT, chemoradiotherapy; DFFR, distant failure–free rate; DFS, disease-free survival; DMFS, disease metastasis–free survival; FFS, failure-free survival; FU, fluorouracil; HR, hazard ratio; LRFFR,

Downloaded from ascopubs.org by University OF HONG KONG Libraries on August 14, 2018 from 147.008.031.043
locoregional failure–free rate; LRS, locoregional relapse free survival; NS, not significant; OS, overall survival; PFS, progression-free survival; RFS, relapse-free survival; RT, radiotherapy.

For concurrent CRT over RT alone.
†Conventional fractionation.
‡Accelerated fractionation.
§Multivariable analysis.

© 2015 by American Society of Clinical Oncology


3359
Lee et al

made regarding the use of adjuvant chemotherapy after CRT in lo- mation consists of a series of seminal events, which include EBV latent
coregionally advanced NPC. The current focus of research is on iden- infection, evasion of host immune surveillance, loss of heterogeneity
tifying patient subgroups that may maximally benefit from adjuvant at specific chromosomal regions, genetic mutations and activation of
chemotherapy. Because elevated level of plasma EBV DNA at 6 to 8 oncogenic signaling pathways, and epigenetic silencing of tumor sup-
weeks after RT could increase the risk of cancer recurrence by almost pressor genes.62-64 In the first genome-wide sequence analysis of NPC
12 times,12 researchers from the Hong Kong NPC study group published recently, Lin et al65 revealed that NPC has a relatively low
(NCT00370890) and NRG Oncology (NCT02135042) are investigat- mutational rate compared with head and neck squamous cell and
ing the utility of post-treatment plasma EBV DNA in individualizing other cancers. Nine significantly mutated genes have been identified as
adjuvant chemotherapy.55a possible driver mutations for NPC, including TP53 and PIK3CA mu-
Available data are relatively limited regarding the role of chemo- tations as well as mutations of genes involved in chromatin transcrip-
therapy for stage II NPC. Chen et al42 randomly assigned 230 patients, tion (BAP1, MLL2, TSHZ3) and cell proliferation (ERBB3, ERBB2,
of whom 87% had stage II and 13% had stage III NPC by AJCC/UICC KRAS, NRAS). The prevalence rates of these mutations are ⱕ 10%.65
(seventh edition) criteria, to RT alone versus CRT. They reported a The ERBB–phosphatidylinositol 3-kinase (PI3K) signaling pathways
significant improvement in OS with CRT over RT alone, with 5-year represent one of the most druggable axes in cancer drug development,
OS rates of 95% versus 86% and an HR of 0.30 (95% CI, 0.12 to 0.76; and genetic alterations (both mutations and amplification) in these
P ⫽ .007). However, the fact that two-dimensional RT was used in this pathways can be found in 23.4% of NPCs.65 The ubiquitous nature of
study suggests that these data should be interpreted in light of the now type II latent EBV infection in nonkeratinizing NPC has also provided
broader application of IMRT. The 5-year disease-specific survival an impetus for investigating EBV-directed therapies, given the knowl-
rates in stage I to II NPC are now expected to be 94% to 97% with edge that activated CD8⫹ T cells are capable of attacking tumor cells
IMRT alone in some retrospective series.21,56 In the study by Su et al,56 and inducing tumor regression in NPC models.66
where 198 patients with stage I to II NPC were treated with IMRT On the basis of this insight into the molecular biology of NPC, the
alone, the 5-year distant metastasis–free rates for T2N0, T1N1, and development of novel therapies has largely followed a three-pronged
T2N1 disease were 98.8%, 100%, and 93.8%, respectively. Given these approach: targeting of signal transduction and angiogenesis, modula-
excellent results with IMRT alone and the added toxicities of concur- tion of gene expression, and cancer immunotherapy (Fig 1). Scientific
rent chemotherapy, such as late hearing loss,21 it is debatable whether progress in these fields has somewhat lagged behind the achievements
CRT should be offered routinely to patients who are undergoing in basic research, and the best evidence in contemporary literature is
IMRT for stage II NPC. Chemotherapy should be confined to patients largely limited to exploratory phase II studies. In the palliative and
with high-risk stage II disease with N1 classification and/or parapha- platinum-refractory populations, studies of epidermal growth factor
ryngeal tumor extension; studies investigating other risk factors, such receptor inhibitors (eg, gefitinib and cetuximab) and angiogenesis
as tumor size and plasma EBV DNA level, are also warranted in the inhibitors (eg, pazopanib and sunitinib) have either reported a lack of
future. For instance, up to one third of patients with stage IIB disease activity or failed to meet their predefined levels of drug efficacy.67,68
with a high pretreatment level of plasma EBV DNA level may develop For instance, in the studies of pazopanib and sunitinib, although the
distant failure.57 median time to progression of 4.4 months seemed to exceed that
reported with cetuximab plus carboplatin (2.7 months),67,68 the me-
dian time to progression in phase II studies of gemcitabine or capecit-
MANAGEMENT OF RECURRENCE AND DEVELOPMENT OF NEW abine alone in similar populations was 5 to 7.5 months.69-71 Likewise,
SYSTEMIC THERAPIES in studies of locoregionally advanced NPC, the addition of cetuximab
or bevacizumab to CRT resulted in survival figures that were similar to
Even with the best available treatment in modern practice, retrospec- or marginally better than historical data on CRT alone.72,73 Another
tive reports of patients treated with IMRT over the last decade have less common reason for failure in phase II studies was an increased
revealed the stark reality that 5% to 15% of patients will develop local incidence of drug-related toxicities, such as tumor bleeding with
failure, and 15% to 30% will experience failure at distant sites.15-24 sunitinib and enhanced RT-related acute mucocutaneous toxicities
Despite the varying success of surgery or reirradiation in salvage ther- with cetuximab.68,73 Perhaps one of the most fundamental barriers to
apy for highly selected patients with local recurrence, a vast majority of the development of targeted therapies for NPC lies in the preclinical
cases of recurrent disease are only amenable to palliative chemother- stage, where there is a scarcity of authentic nonkeratinizing NPC
apy. In phase II studies of platinum-based doublets that are in popular models that are exploitable for demonstrating proof of principle of
use today, the median OS rates in the first-line setting ranged from a novel therapeutic approaches.74-76 To date, only the cell line C666-1
minimum of 11 to 28 months with regimens containing paclitaxel, and a few patient-derived xenografts that were established in the 1990s
fluorouracil, gemcitabine, or capecitabine.58-60 These figures need to have been shown to stably express the EBV genome in culture.75-79 In
be interpreted with caution, because selection bias is inherent in these a recent study using RNA sequencing analysis, it was speculated that
small single-armed studies, and standards of supportive care have some of the cell lines commonly used in NPC studies could have been
changed over time. In contrast, the median time to progression in such contaminated by human papilloma virus– carrying HeLa cells.74 This
studies has remained static at approximately 7.3 to 10 months.58-60 shortage of reliable NPC models, especially those that can stably ex-
Prolonged survival beyond 5 years has been described only for a press certain actionable genetic aberrations in culture, has also con-
minority of younger patients with oligometastases.61 Therefore, the tributed to the difficulty of identifying predictive markers of drug
use of chemotherapy has reached a therapeutic ceiling in NPC. efficacy. This problem has practical implications when designing clin-
It has long been proposed that the molecular pathogenesis of ical trials for drugs against ERBB-PI3K signaling in NPC.65 It is well
EBV-associated NPC follows a stepwise progression.62 This transfor- known that in other cancers, such as head and neck squamous cell

3360 © 2015 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

Downloaded from ascopubs.org by University OF HONG KONG Libraries on August 14, 2018 from 147.008.031.043
Copyright © 2018 American Society of Clinical Oncology. All rights reserved.
Management of Nasopharyngeal Carcinoma

Cetuximab Bevacizumab
Tumor PD-1
VEGF antibody
vasculature
PD-1R
EGFR VEGFR
Gefitinib Sunitinib
Pazopanib

PTEN
Isoform-specific
PI3K inhibitor TIL

PI3K RAS CD8+


AZA T-cell

CD4+

LMP1/2
T-cell
MK2206
inhibitor

AKT RAF
Dual

APC
HAT

EBNA1
Vaccine
AC AC
mTOR MEK

Auto
MEK inhibitor SAHA T-killer

Fig 1. Systemic strategies (noncytotoxic chemotherapy) for advanced nasopharyngeal carcinoma. Drug names in squares have been evaluated in clinical trials. AC,
acetylase; APC, antigen-presenting cells; auto T-killer, autologous cytotoxic T cell; AZA, azacitadine; EBNA1, Epstein-Barr nuclear antigen-1; EGFR, epidermal growth
factor receptor; HAT, histone acetlyase; LMP1/2, latent membrane proteins 1 and 2; mTOR, mammalian target of rapamycin; PD-1R, programmed death-1 receptor;
PI3K, phosphatidylinositol 3-kinase; SAHA, suberoylanilide hydroxamic acid; TIL, tumor-infiltrating lymphocyte; VEGF, vascular endothelial growth factor; VEGFR,
vascular endothelial growth factor receptor.

cancer, tumors that harbor PIK3CA mutations are more sensitive to mors,85 the relatively lower prevalence of T cells found in patients with
PI3K inhibitors than those that do not.80 With the relatively low NPC who could recognize HLA-restricted epitopes in LMP2 and
prevalence of PIK3CA mutations in NPC (⬍ 6%),65 one would need EBNA,86 and the inactivation of tumor infiltrating T cells by the
convincing preclinical data to support the decision of whether to suppressive tumor microenvironment all add to the challenges of
restrict study recruitment based on PIK3CA mutation status. How- developing EBV-directed therapies.86 Successive generations of clini-
ever, the literature is mixed with regard to whether PIK3CA mutations cal studies have tried to boost anti-EBV responses in circulating
could reliably predict response to PI3K–mammalian target of rapa- CD8⫹ T cells in patients with NPC through autologous T-cell
mycin inhibitors in existing models of NPC.81,82 Therefore, a more transfer87-91 or vaccination with dendritic cells or peptides.92-95 Stud-
systematic approach to drug development is needed, and emphasis ies of T-cell therapy have shown that such strategies are generally safe
should be placed on authenticating existing preclinical models, over- and effective in inducing LMP2-specific immune responses, and du-
coming the technical difficulties of establishing new patient-derived rable tumor regressions in advanced NPC have been reported in
xenograft models of NPC, and adopting a more biomarker-based earlier studies.88,89 Recent studies have focused on sequencing adop-
approach to developing new drugs. tive T-cell therapy with chemotherapy in advanced NPC91 and on
Cancer immunotherapy represents another pillar of therapeutic optimizing T-cell expansion using novel adenovirus vectors.90 Using a
approaches in NPC. This encompasses a broad range of strategies, non– cell-based approach, Hui et al95 reported their experience with a
including EBV-directed adoptive and active immunotherapies, anti- recombinant vaccinia virus– based vaccine, which encodes a function-
bodies, EBV lytic cycle induction, and immune-checkpoint blockade. ally inactive fusion protein containing the CD4 epitope–rich
The cardinal feature of EBV infection in NPC is that EBV expresses a C-terminal half of EBNA1 and CD8 epitope–rich LMP2A. This potent
set of poorly immunogenic latent type II viral antigens. EB nuclear vaccine could induce T-cell response in ⬎ 80% of patients, in some
antigen-1 (EBNA1) is expressed frequently in NPC and is a dominant cases boosting response to both CD4⫹- and CD8⫹-mediated immu-
target for CD4⫹ T cells. Latent membrane protein-1 (LMP1) and nity against EBNA1 and/or LMP2.95 This vaccine is being evaluated in
LMP2 are expressed in approximately 50% of NPC tumors, and both a phase II trial involving patients who have detectable plasma EBV
are targets for CD8⫹ cytotoxic T lymphocytes (CTLs). Because LMP1 DNA after RT or who experience optimal response to palliative che-
is poorly immunogenic, LMP2 is considered a more likely target motherapy (NCT01094405).
antigen for CD8⫹ CTL-based therapy.83 It has been suggested that Despite the progress in immunotherapeutic approaches for
NPC cells have preserved antigen-processing function and can be NPC, these strategies remain experimental, and implementation is
recognized by major histocompatibility complex class I–restricted difficult in clinical practice because of their expense and reliance on
virus-specific CTLs in vitro.84 However, the downregulation of major specialized centers. Recent breakthroughs in the treatment of mela-
histocompatibility complex class I peptide expression in NPC tu- noma and other cancers with immune-checkpoint inhibitors have

www.jco.org © 2015 by American Society of Clinical Oncology 3361

Downloaded from ascopubs.org by University OF HONG KONG Libraries on August 14, 2018 from 147.008.031.043
Copyright © 2018 American Society of Clinical Oncology. All rights reserved.
Lee et al

rejuvenated enthusiasm in immunotherapy for NPC. Programmed institutions in the development of new NPC models, the high-
cell death ligand-1 (PD-L1) is highly expressed by cancer cells and throughput screening of new compounds in NPC models through
tumor-infiltrating macrophages in virus-associated malignancies engagement of the interests of pharmaceutical companies, and the
such as NPC,96 and there is emerging evidence that activation of the rationalization of clinical trial design based on unmet clinical needs
PD-1 pathway may contribute to immune escape in NPC models.97,98 and preclinical proof-of-principle studies. The concept of individual-
PD-L1 is expressed in up to 90% of NPC tumors,96,97 and because ized therapy also applies to the optimal selection of patients for ad-
there is now evidence to suggest that PD-L1 expression may predict junctive chemotherapy during RT, and multinational studies are now
response to PD-L1 antibodies in some cancers, there is sufficient addressing the role of adjuvant chemotherapy in subpopulations with
rationale to initiate studies of PD-L1 inhibitors and related agents elevated plasma EBV DNA levels after RT. With the expanding arsenal
in NPC. and diversity of systemic agents on the horizon, it is only a matter of
time before researchers are faced with the dilemma of how to opti-
mally integrate these agents into conventional therapeutic algorithms.
DISCUSSION More well-designed multi-institutional studies are needed, and this
calls for a collective effort among expert centers in harmonizing and
The current overview has highlighted the achievements and challenges standardizing procedures such as biomarker analysis, disease staging,
in the contemporary management of NPC, and insight into these and treatment planning.
issues will set the goals and directions for future research. These goals
maybe thematically summed up as follow: improvement in individu- AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS
alized therapy through better precision in selecting and delivering OF INTEREST
treatments, improvement in the integration of multimodal therapies,
and interdisciplinary cooperation and cross-institutional collabora- Disclosures provided by the authors are available with this article at
tion. The precision of RT planning and delivery could potentially be www.jco.org.
refined in individuals through the use of adaptive RT, image-guided
RT, and proton therapy. Likewise, with the discovery of new AUTHOR CONTRIBUTIONS
driver mutations, putative tumor suppressor genes, and immune-
checkpoint targets in NPC, it is time to adopt a more systematic Conception and design: Anne W.M. Lee, Anthony T.C. Chan
approach to new drug development. Emphasis should be placed on Manuscript writing: All authors
the promotion of interdisciplinary collaboration within and across Final approval of manuscript: All authors

9. Wan XB, Wei L, Li H, et al: High pretreatment 17. Lin S, Pan J, Han L, et al: Update report of
REFERENCES serum lactate dehydrogenase level correlates with nasopharyngeal carcinoma treated with reduced-
disease relapse and predicts an inferior outcome in volume intensity-modulated radiation therapy and
1. National Comprehensive Cancer Network: locally advanced nasopharyngeal carcinoma. Eur J hypothesis of the optimal margin. Radiother Oncol
NCCN treatment guidelines in oncology: Head and neck Cancer 49:2356-2364, 2013 110:385-389, 2014
cancers, 2014. http://www.nccn.org/professionals/ 10. Lo YM, Chan AT, Chan LY, et al: Molecular 18. Wu F, Wang R, Lu H, et al: Concurrent che-
physician_gls/f_guidelines.asp prognostication of nasopharyngeal carcinoma by moradiotherapy in locoregionally advanced nasopha-
2. Chan AT, Grégoire V, Lefebvre JL, et al: quantitative analysis of circulating Epstein-Barr virus ryngeal carcinoma: Treatment outcomes of a
Nasopharyngeal cancer: EHNS-ESMO-ESTRO clini- DNA. Cancer Res 60:6878-6881, 2000 prospective, multicentric clinical study. Radiother
cal practice guidelines for diagnosis, treatment and 11. Lo YM, Chan LY, Lo KW, et al: Quantitative Oncol 112:106-111, 2014
follow-up. Ann Oncol 23:vii83-vii85, 2012 (suppl 7) analysis of cell-free Epstein-Barr virus DNA in 19. Sun X, Su S, Chen C, et al: Long-term out-
3. Ho JHC: Stage classification of nasopharyn- plasma of patients with nasopharyngeal carcinoma. comes of intensity-modulated radiotherapy for 868
geal carcinoma, in de The G and Ito Y (eds): Naso- Cancer Res 59:1188-1191, 1999 patients with nasopharyngeal carcinoma: An analy-
pharyngeal Carcinoma, Etiology and Control. Lyon, 12. Chan AT, Lo YM, Zee B, et al: Plasma Epstein- sis of survival and treatment toxicities. Radiother
France, IARC Scientific Publications, 1978, pp 99- Barr virus DNA and residual disease after radiother- Oncol 110:398-403, 2014
113 apy for undifferentiated nasopharyngeal carcinoma. 20. Ng WT, Lee MC, Chang AT, et al: The impact
4. Ng WT, Lee AW, Kan WK, et al: N-staging by J Natl Cancer Inst 94:1614-1619, 2002 of dosimetric inadequacy on treatment outcome of
magnetic resonance imaging for patients with naso- 13. Liu N, Chen NY, Cui RX, et al: Prognostic value nasopharyngeal carcinoma with IMRT. Oral Oncol
pharyngeal carcinoma: Pattern of nodal involvement of a microRNA signature in nasopharyngeal carci- 50:506-512, 2014
by radiological levels. Radiother Oncol 82:70-75, noma: A microRNA expression analysis. Lancet On- 21. Lee AW, Ng WT, Chan LL, et al: Evolution of
2007 col 13:633-641, 2012 treatment for nasopharyngeal cancer: Success and
5. Yue D, Xu YF, Zhang F, et al: Is replacement 14. Wang HY, Sun BY, Zhu ZH, et al: Eight- setback in the intensity-modulated radiotherapy era.
of the supraclavicular fossa with the lower level signature classifier for prediction of nasopharyngeal Radiother Oncol 11:377-384, 2014
classification based on magnetic resonance imaging carcinoma survival. J Clin Oncol 29:4516-4525, 2011 22. Yi J, Huang X, Gao L, et al: Intensity-modulated
beneficial in nasopharyngeal carcinoma? Radiother 15. Lai SZ, Li WF, Chen L, et al: How does radiotherapy with simultaneous integrated boost for lo-
Oncol 113:108-114, 2014 intensity-modulated radiotherapy versus conven- coregionally advanced nasopharyngeal carcinoma. Radiat
6. Lee AW, Lin JC, Ng WT: Current manage- tional two-dimensional radiotherapy influence the Oncol 9:56, 2014
ment of nasopharyngeal cancer. Semin Radiat Oncol treatment results in nasopharyngeal carcinoma pa- 23. Kam MK, Leung SF, Zee B, et al: Prospective
22:233-244, 2012 tients? Int J Radiat Oncol Biol Phys 80:661-668, randomized study of intensity-modulated radiother-
7. Sze H, Chan LL, Ng WT, et al: Should all 2011 apy on salivary gland function in early-stage naso-
nasopharyngeal carcinoma with masticator space 16. Peng G, Wang T, Yang KY, et al: A prospec- pharyngeal carcinoma patients. J Clin Oncol 25:
involvement be staged as T4? Oral Oncol 50:1188- tive, randomized study comparing outcomes and 4873-4879, 2007
1195, 2014 toxicities of intensity-modulated radiotherapy vs. 24. Pow EH, Kwong DL, McMillan AS, et al:
8. Ng WT, Yuen KT, Au KH, et al: Staging of conventional two-dimensional radiotherapy for the Xerostomia and quality of life after intensity-
nasopharyngeal carcinoma: The past, the present treatment of nasopharyngeal carcinoma. Radiother modulated radiotherapy vs. conventional radiother-
and the future. Oral Oncol 50:549-554, 2014 Oncol 104:286-293, 2012 apy for early-stage nasopharyngeal carcinoma: Initial

3362 © 2015 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

Downloaded from ascopubs.org by University OF HONG KONG Libraries on August 14, 2018 from 147.008.031.043
Copyright © 2018 American Society of Clinical Oncology. All rights reserved.
Management of Nasopharyngeal Carcinoma

report on a randomized controlled clinical trial. Int J therapy alone in patients with stage III to IVB 53. Lee AW, Ngan RK, Tung SY, et al: Preliminary
Radiat Oncol Biol Phys 66:981-991, 2006 nasopharyngeal carcinoma from endemic regions of results of trial NPC-0501 evaluating the therapeutic
25. Gao Y, Zhu G, Lu J, et al: Is elective irradiation China. Cancer 119:2230-2238, 2013 gain by changing from concurrent-adjuvant to
to the lower neck necessary for N0 nasopharyngeal 40. Lee AW, Tung SY, Ngan RK, et al: Factors induction-concurrent chemoradiotherapy, changing
carcinoma? Int J Radiat Oncol Biol Phys 77:1397- contributing to the efficacy of concurrent-adjuvant from fluorouracil to capecitabine, and changing from
1402, 2010 chemotherapy for locoregionally advanced nasopha- conventional to accelerated radiotherapy fraction-
26. Li JG, Yuan X, Zhang LL, et al: A randomized ryngeal carcinoma: Combined analyses of NPC-9901 ation in patients with locoregionally advanced naso-
clinical trial comparing prophylactic upper versus and NPC-9902 trials. Eur J Cancer 47:656-666, 2011 pharyngeal carcinoma. Cancer 121:1328-1338, 2015
whole-neck irradiation in the treatment of patients 41. Lee AW, Tung SY, Chua DT, et al: Random- 54. Yen RF, Chen TH, Ting LL, et al: Early restag-
with node-negative nasopharyngeal carcinoma. Can- ized trial of radiotherapy plus concurrent-adjuvant ing whole-body (18)F-FDG PET during induction che-
cer 119:3170-3176, 2013 chemotherapy vs radiotherapy alone for regionally motherapy predicts clinical outcome in patients with
27. Lee N, Harris J, Garden AS, et al: Intensity- advanced nasopharyngeal carcinoma. J Natl Cancer locoregionally advanced nasopharyngeal carcinoma.
modulated radiation therapy with or without chemo- Inst 102:1188-1198, 2010 Eur J Nucl Med Mol Imaging 32:1152-1159, 2005
therapy for nasopharyngeal carcinoma: Radiation 42. Chen QY, Wen YF, Guo L, et al: Concurrent 55. Chen L, Hu CS, Chen XZ, et al: Concurrent
Therapy Oncology Group phase II trial 0225. J Clin chemoradiotherapy vs radiotherapy alone in stage II chemoradiotherapy plus adjuvant chemotherapy
Oncol 27:3684-3690, 2009 nasopharyngeal carcinoma: Phase III randomized versus concurrent chemoradiotherapy alone in pa-
28. Cheng HC, Wu VW, Ngan RK, et al: A pro- trial. J Natl Cancer Inst 103:1761-1770, 2011 tients with locoregionally advanced nasopharyngeal
spective study on volumetric and dosimetric 43. Chan AT, Teo PM, Ngan RK, et al: Concurrent carcinoma: A phase 3 multicentre randomised con-
changes during intensity-modulated radiotherapy for chemotherapy-radiotherapy compared with radiotherapy trolled trial. Lancet Oncol 13:163-171, 2011
nasopharyngeal carcinoma patients. Radiother On- alone in locoregionally advanced nasopharyngeal carci- 55a. Chan ATC, Ngan RKC, NR, Hui EP, et al: A
col 104:317-323, 2012 noma: Progression-free survival analysis of a phase III multicenter randomized controlled trial (RCT) of ad-
29. Wang X, Lu J, Xiong X, et al: Anatomic and randomized trial. J Clin Oncol 20:2038-2044, 2002 juvant chemotherapy (CT) in nasopharyngeal carci-
dosimetric changes during the treatment course of 44. Lin JC, Jan JS, Hsu CY, et al: Phase III study noma (NPC) with residual plasma EBV DNA (EBV
intensity-modulated radiotherapy for locally ad- of concurrent chemoradiotherapy versus radiother- DNA) following primary radiotherapy (RT) or chemo-
vanced nasopharyngeal carcinoma. Med Dosim 35: apy alone for advanced nasopharyngeal carcinoma: radiotherapy (CRT). J Clin Oncol 30:358s, 2012
151-157, 2010 Positive effect on overall and progression-free sur- (suppl 15s; abstr 5511)
30. Wang W, Yang H, Hu W, et al: Clinical study of vival. J Clin Oncol 21:631-637, 2003 56. Su SF, Han F, Zhao C, et al: Long-term out-
the necessity of replanning before the 25th fraction 45. Lee AW, Tung SY, Chan AT, et al: Preliminary comes of early-stage nasopharyngeal carcinoma pa-
during the course of intensity-modulated radiother-
results of a randomized study (NPC-9902 Trial) on tients treated with intensity-modulated radiotherapy
apy for patients with nasopharyngeal carcinoma. Int
therapeutic gain by concurrent chemotherapy and/or alone. Int J Radiat Oncol Biol Phys 82:327-333, 2012
J Radiat Oncol Biol Phys 77:617-621, 2010
accelerated fractionation for locally advanced naso- 57. Leung SF, Chan AT, Zee B, et al: Pretherapy
31. Yang H, Hu W, Wang W, et al: Replanning
pharyngeal carcinoma. Int J Radiat Oncol Biol Phys quantitative measurement of circulating Epstein-
during intensity modulated radiation therapy im-
66:142-151, 2006 Barr virus DNA is predictive of posttherapy distant
proved quality of life in patients with nasopharyngeal
46. Lee AW, Lau WH, Tung SY, et al: Preliminary failure in patients with early-stage nasopharyngeal
carcinoma. Int J Radiat Oncol Biol Phys 85:e47-e54,
results of a randomized study on therapeutic gain by carcinoma of undifferentiated type. Cancer 98:288-
2013
concurrent chemotherapy for regionally-advanced 291, 2003
32. Lee AW, Lau KY, Hung WM, et al: Potential
nasopharyngeal carcinoma: NPC-9901 trial by the 58. Ngan RK, Yiu HH, Lau WH, et al: Combination
improvement of tumor control probability by induc-
Hong Kong Nasopharyngeal Cancer Study Group. J gemcitabine and cisplatin chemotherapy for meta-
tion chemotherapy for advanced nasopharyngeal
Clin Oncol 23:6966-6975, 2005 static or recurrent nasopharyngeal carcinoma: Re-
carcinoma. Radiother Oncol 87:204-210, 2008
47. Zhang L, Zhao C, Peng PJ, et al: Phase III port of a phase II study. Ann Oncol 13:1252-1258,
33. Chan A, Adams AJ, Weyman E, et al: A phase
study comparing standard radiotherapy with or with- 2002
II trial of proton radiation therapy with chemotherapy
out weekly oxaliplatin in treatment of locoregionally 59. Tan EH, Khoo KS, Wee J, et al: Phase II trial of
for nasopharyngeal carcinoma. Int J Radiat Oncol
advanced nasopharyngeal carcinoma: Preliminary a paclitaxel and carboplatin combination in Asian
Biol Phys 84:S151-S152, 2012
results. J Clin Oncol 23:8461-8468, 2005 patients with metastatic nasopharyngeal carcinoma.
34. Lee AW, Sze WM, Au JS, et al: Treatment
results for nasopharyngeal carcinoma in the modern 48. Loong HH, Ma BB, Leung SF, et al: Prognostic Ann Oncol 10:235-237, 1999
era: The Hong Kong experience. Int J Radiat Oncol significance of the total dose of cisplatin adminis- 60. Chua DT, Yiu HH, Seetalarom K, et al: Phase II
Biol Phys 61:1107-1116, 2005 tered during concurrent chemoradiotherapy in pa- trial of capecitabine plus cisplatin as first-line therapy
35. Wu X, Huang PY, Peng PJ, et al: Long-term tients with locoregionally advanced nasopharyngeal in patients with metastatic nasopharyngeal cancer.
follow-up of a phase III study comparing radiother- carcinoma. Radiother Oncol 104:300-304, 2012 Head Neck 34:1225-1230, 2012
apy with or without weekly oxaliplatin for locore- 49. Blanchard P, Lee AWM, Leclercq J, et al: 61. Teo PM, Kwan WH, Lee WY, et al: Prognos-
gionally advanced nasopharyngeal carcinoma. Ann Meta-analysis of chemotherapy in nasopharyngeal ticators determining survival subsequent to distant
Oncol 24:2131-2136, 2013 carcinoma (MAC-NPC): An update on 4,798 pa- metastasis from nasopharyngeal carcinoma. Cancer
36. Chan AT, Leung SF, Ngan RK, et al: Overall tients. J Clin Oncol 32:389s, 2014 (suppl 15s; abstr 77:2423-2431, 1996
survival after concurrent cisplatin-radiotherapy com- 6022) 62. Lo KW, Chung GT, To KF: Deciphering the
pared with radiotherapy alone in locoregionally ad- 50. Hui EP, Ma BB, Leung SF, et al: Randomized molecular genetic basis of NPC through molecular,
vanced nasopharyngeal carcinoma. J Natl Cancer phase II trial of concurrent cisplatin-radiotherapy cytogenetic, and epigenetic approaches. Semin
Inst 97:536-539, 2005 with or without neoadjuvant docetaxel and cisplatin Cancer Biol 22:79-86, 2012
37. Wee J, Tan EH, Tai BC, et al: Randomized trial in advanced nasopharyngeal carcinoma. J Clin Oncol 63. Lo KW, Huang DP: Genetic and epigenetic
of radiotherapy versus concurrent chemoradiother- 27:242-249, 2009 changes in nasopharyngeal carcinoma. Semin Can-
apy followed by adjuvant chemotherapy in patients 51. Tan T, Lim WT, Fong KW, et al: Randomized cer Biol 12:451-462, 2002
with American Joint Committee on Cancer/Interna- phase III trial of concurrent chemoradiation with or 64. Gourzones C, Busson P, Raab-Traub N:
tional Union against cancer stage III and IV nasopha- without neoadjuvant gemcitabine, carboplatin, and Epstein-Barr virus and the pathogenesis of nasopha-
ryngeal cancer of the endemic variety. J Clin Oncol paclitaxel in locally advanced nasopharyngeal can- ryngeal carcinomas, in Busson P (ed): Nasopharyn-
23:6730-6738, 2005 cer. J Clin Oncol 32:384s, 2014 (suppl 15s; abstr geal Carcinoma: Keys for Translational Medicine and
38. Al-Sarraf M, LeBlanc M, Giri PG, et al: Che- 6003) Biology. New York, NY, Landes Bioscience/Springer
moradiotherapy versus radiotherapy in patients with 52. Fountzilas G, Ciuleanu E, Bobos M, et al: Science and Business Media, 2013, pp 42-60
advanced nasopharyngeal cancer: Phase III random- Induction chemotherapy followed by concomitant 65. Lin DC, Meng X, Hazawa M, et al: The
ized Intergroup study 0099. J Clin Oncol 16:1310- radiotherapy and weekly cisplatin versus the same genomic landscape of nasopharyngeal carcinoma.
1317, 1998 concomitant chemoradiotherapy in patients with Nat Genet 46:866-871, 2014
39. Chen Y, Sun Y, Liang SB, et al: Progress nasopharyngeal carcinoma: A randomized phase II 66. Khanna R, Moss D, Gandhi M: Technology
report of a randomized trial comparing long-term study conducted by the Hellenic Cooperative Oncol- insight: Applications of emerging immunotherapeu-
survival and late toxicity of concurrent chemoradio- ogy Group (HeCOG) with biomarker evaluation. Ann tic strategies for Epstein-Barr virus-associated ma-
therapy with adjuvant chemotherapy versus radio- Oncol 23:427-435, 2012 lignancies. Nat Clin Pract Oncol 2:138-149, 2005

www.jco.org © 2015 by American Society of Clinical Oncology 3363

Downloaded from ascopubs.org by University OF HONG KONG Libraries on August 14, 2018 from 147.008.031.043
Copyright © 2018 American Society of Clinical Oncology. All rights reserved.
Lee et al

67. Lim WT, Ng QS, Ivy P, et al: A phase II study harbouring Epstein-Barr virus. Int J Cancer 83:121- 89. Straathof KC, Bollard CM, Popat U, et al:
of pazopanib in Asian patients with recurrent/meta- 126, 1999 Treatment of nasopharyngeal carcinoma with
static nasopharyngeal carcinoma. Clin Cancer Res 78. Busson P, Ganem G, Flores P, et al: Establish- Epstein-Barr virus-specific T lymphocytes. Blood
17:5481-5489, 2011 ment and characterization of three transplantable 105:1898-1904, 2005
68. Hui EP, Ma BB, King AD, et al: Hemorrhagic EBV-containing nasopharyngeal carcinomas. Int J 90. Smith C, Tsang J, Beagley L, et al: Effective
complications in a phase II study of sunitinib in Cancer 42:599-606, 1988 treatment of metastatic forms of Epstein-Barr virus-
patients of nasopharyngeal carcinoma who has pre- 79. Huang DP, Ho JH, Chan WK, et al: Cytogenet- associated nasopharyngeal carcinoma with a novel
viously received high-dose radiation. Ann Oncol 22: ics of undifferentiated nasopharyngeal carcinoma adenovirus-based adoptive immunotherapy. Cancer
1280-1287, 2011 xenografts from southern Chinese. Int J Cancer Res 72:1116-1125, 2012
69. Foo KF, Tan EH, Leong SS, et al: Gemcitabine 43:936-939, 1989 91. Chia WK, Teo M, Wang WW, et al: Adoptive
in metastatic nasopharyngeal carcinoma of the un- 80. Lui VW, Hedberg ML, Li H, et al: Frequent T-cell transfer and chemotherapy in the first-line
differentiated type. Ann Oncol 13:150-156, 2002 mutation of the PI3K pathway in head and neck treatment of metastatic and/or locally recurrent na-
70. Chua DT SJ, Au GK: A phase II study of cancer defines predictive biomarkers. Cancer Dis- sopharyngeal carcinoma. Mol Ther 22:132-139,
capecitabine in patients with recurrent and meta- cov 3:761-769, 2013 2014
static nasopharyngeal carcinoma pretreated with 81. Yang F, Qian XJ, Qin W, et al: Dual phospho- 92. Lin CL, Lo WF, Lee TH, et al: Immunization
platinum-based chemotherapy. Oral Oncol 39:361- inositide 3-kinase/mammalian target of rapamycin with Epstein-Barr Virus (EBV) peptide-pulsed den-
366, 2004 inhibitor NVP-BEZ235 has a therapeutic potential dritic cells induces functional CD8⫹ T-cell immunity
71. Zhang L, Zhang Y, Huang PY, et al: Phase II and sensitizes cisplatin in nasopharyngeal carci- and may lead to tumor regression in patients with
noma. PLoS One 8:e59879, 2013 EBV-positive nasopharyngeal carcinoma. Cancer
clinical study of gemcitabine in the treatment of
82. Ma BB, Lui VW, Hui CW, et al: Preclinical Res 62:6952-6958, 2002
patients with advanced nasopharyngeal carcinoma
evaluation of the mTOR-PI3K inhibitor BEZ235 in 93. Chia WK, Wang WW, Teo M, et al: A phase II
after the failure of platinum-based chemotherapy.
nasopharyngeal cancer models. Cancer Lett 343:24- study evaluating the safety and efficacy of an
Cancer Chemother Pharmacol 61:33-38, 2008
32, 2014 adenovirus-DeltaLMP1-LMP2 transduced dendritic
72. Lee NY, Zhang Q, Pfister DG, et al: Addition of
83. Lee SP: Nasopharyngeal carcinoma and the cell vaccine in patients with advanced metastatic
bevacizumab to standard chemoradiation for locore-
EBV-specific T cell response: Prospects for immu- nasopharyngeal carcinoma. Ann Oncol 23:997-1005,
gionally advanced nasopharyngeal carcinoma (RTOG
notherapy. Semin Cancer Biol 12:463-471, 2002 2012
0615): A phase 2 multi-institutional trial. Lancet
84. Khanna R, Busson P, Burrows SR, et al: 94. Li F, Song D, Lu Y, et al: Delayed-type hyper-
Oncol 13:172-180, 2011
Molecular characterization of antigen-processing sensitivity (DTH) immune response related with
73. Ma BB, Kam MK, Leung SF, et al: A phase II
function in nasopharyngeal carcinoma (NPC): Evi- EBV-DNA in nasopharyngeal carcinoma treated with
study of concurrent cetuximab-cisplatin and dence for efficient presentation of Epstein-Barr virus autologous dendritic cell vaccination after radiother-
intensity-modulated radiotherapy in locoregionally cytotoxic T-cell epitopes by NPC cells. Cancer Res apy. J Immunother 36:208-214, 2013
advanced nasopharyngeal carcinoma. Ann Oncol 58:310-314, 1998 95. Hui EP, Taylor GS, Jia H, et al: Phase 1 trial of
23:1287-1292, 2011 85. Yao Y, Minter HA, Chen X, et al: Heterogene- recombinant modified vaccinia ankara (MVA) encod-
74. Strong MJ, Baddoo M, Nanbo A, et al: Compre- ity of HLA and EBER expression in Epstein-Barr ing Epstein-Barr viral tumor antigens in nasopharyn-
hensive high-throughput RNA sequencing analysis virus-associated nasopharyngeal carcinoma. Int J geal carcinoma patients. Cancer Res 73:1676-1688,
reveals contamination of multiple nasopharyngeal carci- Cancer 88:949-955, 2000 2013
noma cell lines with HeLa cell genomes. J Virol 88: 86. Li J, Zeng XH, Mo HY, et al: Functional 96. Chen BJ, Chapuy B, Ouyang J, et al: PD-L1
10696-10704, 2014 inactivation of EBV-specific T-lymphocytes in naso- expression is characteristic of a subset of aggres-
75. Gullo C, Low WK, Teoh G: Association of pharyngeal carcinoma: Implications for tumor immu- sive B-cell lymphomas and virus-associated malig-
Epstein-Barr virus with nasopharyngeal carcinoma notherapy. PLoS One 2:e1122, 2007 nancies. Clin Cancer Res 19:3462-3473, 2013
and current status of development of cancer-derived 87. Chua D, Huang J, Zheng B, et al: Adoptive 97. Hsu MC, Hsiao JR, Chang KC, et al: Increase
cell lines. Ann Acad Med Singapore 37:769-777, transfer of autologous Epstein-Barr virus-specific of programmed death-1-expressing intratumoral
2008 cytotoxic T cells for nasopharyngeal carcinoma. Int J CD8 T cells predicts a poor prognosis for nasopha-
76. Chan SY, Choy KW, Tsao SW, et al: Authen- Cancer 94:73-80, 2001 ryngeal carcinoma. Mod Pathol 23:1393-1403, 2010
tication of nasopharyngeal carcinoma tumor lines. 88. Comoli P, Pedrazzoli P, Maccario R, et al: Cell 98. Fang W, Zhang J, Hong S, et al: EBV-driven
Int J Cancer 122:2169-2171, 2008 therapy of stage IV nasopharyngeal carcinoma with LMP1 and IFN-gamma up-regulate PD-L1 in naso-
77. Cheung ST, Huang DP, Hui AB, et al: Naso- autologous Epstein-Barr virus-targeted cytotoxic T pharyngeal carcinoma: Implications for oncotar-
pharyngeal carcinoma cell line (C666-1) consistently lymphocytes. J Clin Oncol 23:8942-8949, 2005 geted therapy. Oncotarget 5:12189-12202, 2014

■ ■ ■

3364 © 2015 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

Downloaded from ascopubs.org by University OF HONG KONG Libraries on August 14, 2018 from 147.008.031.043
Copyright © 2018 American Society of Clinical Oncology. All rights reserved.
Management of Nasopharyngeal Carcinoma

AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

Management of Nasopharyngeal Carcinoma: Current Practice and Future Perspective


The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated. Relationships are
self-held unless noted. I ⫽ Immediate Family Member, Inst ⫽ My Institution. Relationships may not relate to the subject matter of this manuscript. For more
information about ASCO’s conflict of interest policy, please refer to www.asco.org/rwc or jco.ascopubs.org/site/ifc.
Anne W.M. Lee Anthony T.C. Chan
No relationship to disclose Consulting or Advisory Role: Merck, Taiho Pharmaceutical, Roche,
Amgen
Brigette B.Y. Ma
Research Funding: Boehringer Ingelheim, Bristol-Myers Squibb, Eli
Honoraria: Novartis, Bayer, Boehringer Ingelheim
Consulting or Advisory Role: Novartis, Bayer, Boehringer Ingelheim Lilly, Pfizer
Research Funding: Novartis
Wai Tong Ng
No relationship to disclose

www.jco.org © 2015 by American Society of Clinical Oncology

Downloaded from ascopubs.org by University OF HONG KONG Libraries on August 14, 2018 from 147.008.031.043
View publication stats Copyright © 2018 American Society of Clinical Oncology. All rights reserved.

You might also like