Sei sulla pagina 1di 17

Seminar

Nasopharyngeal carcinoma
Yu-Pei Chen, Anthony T C Chan, Quynh-Thu Le, Pierre Blanchard, Ying Sun*, Jun Ma*

Lancet 2019; 394: 64–80 Nasopharyngeal carcinoma is characterised by distinct geographical distribution and is particularly prevalent in east
Published Online and southeast Asia. Epidemiological trends in the past decade have shown that its incidence has declined gradually
June 6, 2019 but progressively, and mortality has been reduced substantially. These findings probably reflect lifestyle and
http://dx.doi.org/10.1016/
S0140-6736(19)30956-0
environmental changes, enhanced understanding of the pathogenesis and risk factors, population screening,
advancements in imaging techniques, and individualised comprehensive chemoradiotherapy strategies. In particular,
*Joint senior authors
plasma Epstein-Barr virus (EBV) DNA has been used for population screening, prognostication, predicting treatment
Department of Radiation
Oncology, Sun Yat-sen
response for therapeutic adaptation, and disease surveillance. Moreover, the widespread application of intensity-
University Cancer Center, State modulated radiotherapy and optimisation of chemotherapy strategies (induction, concurrent, adjuvant) have
Key Laboratory of Oncology in contributed to improved survival with reduced toxicities. Among the existing developments in novel therapeutics,
South China, Collaborative
immune checkpoint therapies have achieved breakthroughs for treating recurrent or metastatic disease and represent
Innovation Center for Cancer
Medicine, Guangdong Key a promising future direction in nasopharyngeal carcinoma.
Laboratory of Nasopharyngeal
Carcinoma Diagnosis and Introduction past decades, nasopharyngeal carcinoma incidence has
Therapy, Guangzhou, People’s
Nasopharyngeal carcinoma is an epithelial carcinoma declined gradually worldwide: substantial reductions have
Republic of China (Y-P Chen MD;
Prof Y Sun MD, Prof Jun Ma MD); arising from the nasopharyngeal mucosal lining. In been observed in south and east Asia, north America,
Partner State Key Laboratory of the nasopharynx, the tumour is often observed at and the Nordic countries, with average annual changes of
Oncology in South China, the pharyngeal recess (fossa of Rosenmüller). Despite about –1% to –5%.3,4 In endemic regions, for example,
Sir Y K Pao Centre for Cancer,
originating from similar cell or tissue lineages, naso­ the incidence rate has steadily decreased since the 1980s
Department of Clinical
Oncology, Hong Kong Cancer pharyngeal carcinoma and other epithelial head and neck in Hong Kong, with a total decrease of about 30% over a
Institute and Prince of Wales tumours are distinctly different. 20-year period;5 urban Guangzhou has shown average
Hospital, The Chinese In comparison with other cancers, nasopharyngeal annual changes of about –3% for men and –5% for
University of Hong Kong,
carcinoma is relatively uncommon. According to the women in the 2000–11 period.6 Lifestyle and environmental
Hong Kong, China
(A T C Chan MD); Department of International Agency for Research on Cancer, in 2018, changes may well be the contributory factors.
Radiation Oncology, Stanford there were about 129 000 new cases of nasopharyngeal Nasopharyngeal carcinoma incidence is higher in
University, Stanford, CA, USA carcinoma, accounting for only 0·7% of all cancers males than in females, with a ratio of about 2·5 in
(Q-T Le MD); and Department
of Radiation Oncology,
diagnosed in 2018.1,2 Nevertheless, its geographical global China in 2015.7 It is noteworthy that the high incidence
Gustave-Roussy; Centre for distribution is extremely unbalanced; >70% of new cases in people from southern China remains even after they
Research in Epidemiology and are in east and southeast Asia, with an age-standardised immigrate to non-endemic areas, but reduced incidence
Population Health, INSERM rate (world) of 3·0 per 100 000 in China to 0·4 per 100 000 in has been observed in second-generation migrants.
U1018, Paris-Saclay University,
Villejuif, France
populations that are mainly white (figure 1).1,2 Over the There is also a trend towards decreasing incidence the
(Prof P Blanchard MD) farther the population has immigrated.8 These findings
Correspondence to: suggest that a combination of genetic, ethnic, and
Prof Jun Ma, Department of Search strategy and selection criteria environmental factors might affect nasopharyngeal
Radiation Oncology, Sun Yat-sen carcinoma pathogenesis.
University Cancer Center, State We did an extensive search of MEDLINE and PubMed for
Key Laboratory of Oncology in English-language articles published between Jan 1, 1990,
South China, Collaborative and Jan 31, 2019. The Cochrane Library was also searched for Pathology and risk factors
Innovation Center for Cancer
reviews that had been published between 1990 and 2018. According to the World Health Organization, there are
Medicine, Guangdong Key three pathological subtypes of nasopharyngeal carcinoma:
Laboratory of Nasopharyngeal The search terms included: “nasopharyngeal carcinoma”,
Carcinoma Diagnosis and “nasopharynx cancer”, “epidemiology”, “pathology”, keratinising squamous, non-keratinising, and basaloid
Therapy, 510060, China “genetics”, “Epstein-Barr virus”, “staging”, “imaging”, squamous. Non-keratinising nasopharyngeal carcinoma
majun2@mail.sysu.edu.cn
“positron emission tomography”, “radiotherapy”, can be divided into differentiated and undifferentiated
See Online for appendix
“chemotherapy”, “combined treatment modality”, tumours (appendix p 22).9 The keratinising subtype
“salvage therapy”, “immunotherapy”, “clinical trials”, accounts for less than 20% of cases worldwide, and is
and “meta-analysis”. The search included reports on human relatively rare in endemic areas such as southern China;
studies. We prioritised large, current clinical trials or studies the non-­keratinising subtype constitutes most cases in
for selection. The abstracts of recent pertinent conferences endemic areas (>95%) and is predominantly associated
were also included for the most recent updates. with Epstein-Barr virus (EBV) infection.9–11
We evaluated the relevance of the references that had been The remarkable geographical distribution of naso­
selected, which comprised mainly articles that had been pharyngeal carcinoma incidence has spurred studies
published in the last 5 years; we did not exclude older, on its risk factors, and it is suggested that multiple factors,
original studies that have been referenced widely and are including EBV infection, host genetics, and environmental
greatly respected. factors are contributors in the devel­ opment of naso­
pharyngeal carcinoma. Linkage and association studies

64 www.thelancet.com Vol 394 July 6, 2019


Seminar

ASR (world) per 100 000


≥1·5
0·82 to <1·5
0·50 to <0·82
0·39 to <0·50
0·28 to <0·39
0·19 to <0·28
<0·19
Not applicable
No data

Others 28 200 (21·8%)

USA 2216 (1·7%)

Thailand 2200 (1·7%)


Philippines 2913 (2·3%) China 60 558 (47·7%)
Mediterranean countries* 3792 (2·9%)

India 5086 (4·0%)

Vietnam 6212 (4·9%)

Indonesia 17 992 (14·2%)

Figure 1: Worldwide distribution of nasopharyngeal carcinoma in 2018


(A) Estimated age-standardised incidence rates (ARS; world). (B) Pie chart presents the distribution of the estimated number of new cases for the seven countries in
which nasopharyngeal carcinoma is most commonly reported. Source: Globocan 2018.1,2 ARS=age-standardised rates. *Mediterranean countries include Morocco,
Algeria, Tunisia, Libya, Lebanon, Israel, Turkey, and Greece.

have identified that genetic susceptibility genes increase genetic lesions including loss of the CDKN2A/CDKN2B
nasopharyngeal carcinoma risk. HLA genes residing at locus, CCND1 amplification, TP53 mutation, and muta­
the MHC region on chromosome 6p21 have been widely tions in the PI3K/MAPK signalling pathways, chromatin
recognised as major risk loci conferring nasopharyngeal modification, and DNA repair (appendix p 13).20–23 In
carcinoma risk.12,13 Genome-wide association studies have addition, although the EBV oncoprotein latent membrane
also reported other loci outside the MHC (eg—TNFRSF19 protein 1 (LMP1) constitutively activates NF-κB signalling,
on 13q12, MECOM on 3q26, CDKN2A/2B on 9p21, Li and colleagues22 identified mutual exclusivity among
CLPTM1L/TERT on 5p15l; appendix).14–18 A recent study LMP1 overexpression and NF-κB pathway aberrations,
showed that pathogenic heterozygous germline variants suggesting that both somatic and virus-mediated addic­
of MST1R, which encodes a macrophage-stimulating 1 cell tion to NF-κB signalling contributed to pathogenesis of
surface receptor and is key in host defence against viral nasopharyngeal carcinoma. Appendix pp 23–24 depicts
infection, is strongly associated with early-age onset (age the pathway of significant signalling and transcription
≤20 years) of nasopharyngeal carcinoma,19 which may aid factor alterations according to somatic variants identified
population screening. in the above mentioned studies.20–23
Several recent studies exploring the molecular Besides host genetics, EBV infection is perhaps the
landscape of nasopharyngeal carcinoma have identified most common causal agent of nasopharyngeal carcinoma
crucial genomic changes that stimulate the development (figure 2; appendix). Other potential risk factors identified
and progression of disease: multiple loss-of-function by epidemiological studies include family history of
mutations in the NF-kB–negative regulators, recurrent nasopharyngeal carcinoma (appendix p 25), active and

www.thelancet.com Vol 394 July 6, 2019 65


Seminar

Tumour progression
Tumour formation
Clonal expansion of
EBV-infection cells

Latency II Chemotherapy
LMP1+ and Local recurrence
LMP1– radiotherapy
Normal Precancerous lesions
nasopharyngeal
epithelium Activation of telomerase Inflammation Global
↑Cyclin D1 hyper-methylation
Loss of chr.3p and 9p (inactivation
↓p16, RASSF1A
of RASS1A and CDKN2A) Somatic gene
EBV latent infection alterations in
NFκB pathways
Mutations in
MHC class I
genes
Mutations in P13K/MAPK
and chromatin Distant metastasis
remodelling pathways TP53, RAS or
LMP1 BART-miRNAs other mutations
Invasion and metastasis Potentiate tumour growth
Activate oncogenic signalling pathways Escape immune attack
Anti-apoptosis Anti-apoptosis
Inhibit differentiation of squamous cells EBERs
LMP2 Induce IGF-1 Driver mutations
Promote cell survival Anti-apoptosis
Maintain cell stemness BARF1 EBV
Epithelial–mesenchymal transition Promote cell growth
EBNA1 Tumour cell
Promote cell growth
Maintain EBV episome
Carcinogens
Damage DNA

Figure 2: Theory of the roles of Epstein-Barr virus (EBV) infection and genomic changes in the development of nasopharyngeal carcinoma*
Persistent EBV infection and progressive genomic changes promote clonal evolution of nasopharyngeal carcinoma. Persistent EBV infection in genetically mutated
epithelial cells and the proliferation of infected cells are considered the prerequisites for initiating tumorigenic transformation. Chronic exposure of the nasopharyngeal
mucosa to environmental carcinogens increases DNA damage and leads to somatic genetic changes in the epithelial cells. The activation of telomerase activity on
chromosomes 3p and 9p (chr.3p & 9p) and the accumulation of driving events such as inactivation of the tumour suppressor genes RASSF1A and CDKN2A promote the
immortalisation of histologically normal and/or dysplastic cells, genomic instability, and EBV infection. Furthermore, p16 inactivation and cyclin D1 pathway activation
may contribute to undifferentiated nasopharyngeal epithelial cells, supporting EBV infection. The expression of type II EBV latency gene products, such as LMP1, LMP2,
EBNA1, BART miRNAs, EBERs, and BARF1, alters multiple cellular pathways, promotes cell proliferation, regulates the host microenvironment, and thereby promotes
the clonal expansion of EBV-infected pre-invasive nasopharyngeal epithelial cells. Importantly, EBV facilitates global hypermethylation, consequently inactivating a
variety of cancer-related genes. Moreover, during tumour development, acquired mutations of several negative regulatory factors in the NF-κB signalling pathway alter
the activity of various cancer-related genes. All these conditions enhance tumour heterogeneity. In nasopharyngeal carcinoma progression, the loss of LMP1 may be
associated with the acquisition of additional genetic changes during tumour progression, which compensate for LMP1 function. Further genetic alterations are
acquired during tumour progression. In advanced stages, mutations in the MHC class I genes and in the PI3K/MAPK and chromatin remodelling pathways, and somatic
mutations of TP53, RAS, and other genes may be the driving force of local recurrence and distant metastasis of nasopharyngeal carcinoma cells after conventional
treatment. EBV=Epstein-Barr virus. *The theory was proposed by Kwok-Wai Lo (Department of Anatomical & Cellular Pathology, The Chinese University of Hong Kong,
Hong Kong, China), and the figure was modified with his permission.

passive tobacco smoking, consumption of preserved nasopharyngeal carcinoma screening in asymptomatic


foods and alcohol, and oral hygiene.24–28 participants.29
Encouragingly, a recent prospective screening study
Population screening involving more than 20 000 participants30 showed that
Effective population screening could improve treatment plasma EBV DNA detection was useful for nasopharyn­
outcomes by identifying early-stage disease in patients, geal carcinoma screening, with 97·1% sensi­tivity and
and presents an attractive strategy considering the strained 98·6% specificity. Nasopharyngeal carcinoma was
medical resources in endemic areas such as southern detected significantly earlier, with a higher proportion of
China. Although anti-EBV IgA antibody (eg—early antigen stage I–II disease than in a historical cohort (71% vs
[EA-IgA], anti–EBV capsid antigen [VCA-IgA], anti–EBV 20%), and the outcomes were also better compared with
nuclear antigen 1 [EBNA1-IgA]) serological testing is com­ a historical cohort (3-year progression-free survival
monly used to detect incident nasopharyngeal carcinoma, hazard ratio [HR]: 0·10 [95% CI 0·05–0·18]).30 Plasma
the low sensitivities and specificities impede their use in EBV DNA population screening represents a very

66 www.thelancet.com Vol 394 July 6, 2019


Seminar

Seventh edition Eighth edition


Primary tumour (T)
TX Primary tumour cannot be assessed Primary tumour cannot be assessed
T0 ·· No tumour identified, but there is EBV-positive cervical node(s)
involvement
T1 Nasopharynx, oropharynx, or nasal cavity without Nasopharynx, oropharynx, or nasal cavity without parapharyngeal
parapharyngeal extension extension
T2 Parapharyngeal extension Parapharyngeal extension, adjacent soft tissue involvement
(medial pterygoid, lateral pterygoid, prevertebral muscles)
T3 Bony structures of skull base and/or paranasal sinuses Bony structures (skull base, cervical vertebra) and/or paranasal
sinuses
T4 Intracranial, cranial nerves, hypopharynx, orbit, Intracranial extension, cranial nerve, hypopharynx, orbit, extensive
infratemporal fossa or masticator space soft tissue involvement (beyond the lateral surface of the lateral
pterygoid muscle, parotid gland)
Regional lymph nodes (N)
NX Regional lymph nodes cannot be assessed Regional lymph nodes cannot be assessed
N0 No regional lymph node metastasis No regional lymph node metastasis
N1 Unilateral cervical, unilateral, or bilateral retropharyngeal Unilateral cervical, unilateral, or bilateral retropharyngeal lymph
lymph nodes, above the supraclavicular fossa; ≤6 cm nodes, above the caudal border of cricoid cartilage; ≤6 cm
N2 Bilateral metastasis in lymph node(s), ≤6 cm in greatest Bilateral metastasis in lymph node(s), ≤6 cm in greatest dimension,
dimension, above the supraclavicular fossa above the caudal border of cricoid cartilage
N3a >6 cm in dimension >6 cm and/or below caudal border of cricoid cartilage (regardless of
laterality)
N3b Supraclavicular fossa ··
Distant metastasis (M)
M0 No distant metastasis No distant metastasis
M1 Distant metastasis Distant metastasis
Stage group
I T1 N0 M0 T1 N0 M0
II T2 N0–1 M0, T1 N1 M0, T2 N0–1 M0, T0–1 N1 M0
III T1–3 N2 M0, T3 N0–1 M0 T3 N0–2 M0, T0–2 N2 M0
IVA T4 N0–2 M0 T4 or N3 M0
IVB Any T, N3 M0 Any T, any N, M1
IVC Any T, any N, M1 ··
EBV=Epstein-Barr virus. UICC/AJCC=International Union Against Cancer/American Joint Committee on Cancer. T category: T0 is added for EBV-positive cervical node(s)
involvement despite unidentified primary tumour. Involvement of medial pterygoid, lateral pterygoid, and prevertebral muscles is now staged as T2. In T4, the original
“infratemporal fossa or masticator space” has been replaced by specific description of soft tissue involvement to avoid potential ambiguity. N category: the previous N3b
criterion of “supraclavicular fossa” has been changed to “below caudal border of cricoid cartilage”. N3a and N3b have been merged into a single N3. Stage group: the previous
stages IVA and IVB have been merged into IVA. The previous IVC has been upstaged to IVB.

Table 1: Comparison of the 7th and 8th editions of the UICC/AJCC staging system for nasopharyngeal carcinoma

promising approach for detecting early-stage naso­ the 8th edition in 2016 (from the 7th edition in 2009),
pharyngeal carcinoma (appendix). where primary and nodal disease classification, and the
stage groups were further refined (table 1).31,32
Symptoms and diagnosis As the current anatomy-based staging system is
The clinical symptoms and signs of nasopharyngeal insufficient for predicting prognosis or treatment
carcinoma correlate with the involved anatomical benefits, many studies have assessed whether incor­
regions, and comprehensive head and neck evaluation porating other clinical factors and molecular biomarkers
is indicated in patients with presumed diagnosis of into the system would better predict survival. A recent
nasopharyngeal carcinoma (see appendix for more study33 proposed a nomogram combining pre-treatment
detail). plasma EBV DNA and clinicopathological variables, and
found that it resulted in more accurate prognostic
Staging and prognosis prediction for patients with nasopharyngeal carcinoma.
Nasopharyngeal carcinoma is classified according to the Other biomarkers such as DNA methylation, miRNAs,
International Union Against Cancer/American Joint and mRNAs also have demonstrated their prognostic
Committee on Cancer (UICC/AJCC) TNM (tumour-node- value and potential clinical applications in nasopharyngeal
metastasis) staging system. This system was updated to carcinoma. A six–hypermethylated gene panel34 and a

www.thelancet.com Vol 394 July 6, 2019 67


Seminar

five-miRNA signature35 have been associated with survival parallel: a single-copy sequence for comparing between
in nasopharyngeal carcinoma. A gene expression–based patients, and a multicopy sequence for enhancing
signature was recently proposed as a reliable prognostic the assay sensitivity. Considerations were also made
tool for distant metastasis in nasopharyngeal carcinoma, for adapting new technologies such as next-generation
and might be able to predict which patients might sequencing or digital PCR to improve the assay quanti­
benefit from concurrent chemotherapy as well.36 With tation. At least two laboratories are currently working to
the advances in immunotherapy, the prognostic value optimise the assay, which, once validated, can substantially
of programmed death-1 or programmed death-1 ligand improve the staging system and promote daily use of this
(PD-1/PD-L1) expression in nasopharyngeal carcinoma biomarker in the clinic.
has been explored despite inconsistent conclusions,37–41
possibly due to different immune microenvironment Imaging studies
statuses. High intratumoral and stromal tumour- MRI, CT, and ¹⁸F-fluorodeoxyglucose (¹⁸F-FDG)-PET/CT
infiltrating lymphocytes (TILs) have been associated with are currently the most commonly used imaging
favourable outcomes.42 These immune-related prognostic modalities for nasopharyngeal carcinoma staging and
biomarkers may also reflect immunological heterogeneity radiotherapy. With high soft-tissue resolution, MRI is
and guide future immunotherapy. better for evaluating primary tumour extension and
Notably, the addition of pre-treatment plasma EBV DNA retropharyngeal lymph node metastasis than CT, while
to the 8th edition greatly improved its prognostic they share similar accuracy in detecting cervical lymph
performance.43–45 Future revisions of the TNM staging node metastasis (appendix p 27).51,52 ¹⁸F-FDG-PET/CT
system adapted to incorporate plasma EBV DNA represent performs better than conventional work-up (eg—chest
a promising strategy. Beyond pre-treatment EBV DNA, radiography, abdominal ultrasound, skeletal scintigraphy)
unfavourable EBV DNA response after induction chemo­ for diagnosing distant metastasis, and is also relatively
therapy, midcourse of radiotherapy, or post-treatment is more sensitive and accurate for detecting small cervical
also adverse prognosticators for clinical outcomes.46–48 lymph node metastases and local residual, recurrent
These observations present the possibility of incorporating disease, or both.52–55 Therefore, MRI and ¹⁸F-FDG-PET/CT
EBV DNA for risk-stratified treatment adaptation, based might be preferred for staging in patients with high
on liquid biopsy of biomarker response. Well conducted risk of distant metastasis. Recently, the clinical utility
studies with regular longitudinal EBV DNA measurements of simultaneous whole-body ¹⁸F-FDG-PET/MRI was
during and after treatment are warranted to further assess assessed for staging primary nasopharyngeal carcinoma,
the role of EBV DNA in treatment modification during and showed more accuracy than separate MRI and
active therapy and in tumour surveillance after therapy PET/CT, and could serve as a single-step staging modal­
has been completed. However, although EBV DNA has ity.56 Because ¹⁸F-FDG-PET/CT also provides metabolic
been established as a robust prognostic marker in naso­ parameters, it could represent tumour biology and predict
pharyngeal carcinoma, there is comparatively large treatment outcomes. It has been proposed that the
interlaboratory variability, even for the same assay using metabolic indexes of PET/CT performed before and
identical procedures without harmonisation.49 Accordingly, during radiotherapy are useful prognostic factors for
assay standard­ isation is necessary for measuring this predicting patients with high-risk of regional or distant
biomarker in prospective studies that involve more than metastatic failure.57 Combining ¹⁸F-FDG-PET derived
one par­ticipating laboratory. Notably, Le and colleagues49 parameters and EBV DNA stratifies patients with
have shown that harmonisation is feasible, and have nasopharyngeal carcinoma into different risk subgroups
established a standardised assay that is being used in an better than the conventional TNM system.58
international biomarker-driven clinical trial. In addition to The emerging radiomics methods aim to convert
the tech­nical aspects, other challenges in plasma EBV medical images into high-dimensional, quantitative
DNA measurements include the variable number of viral imaging features to improve oncological decision support
episomes within each nasopharyngeal carcinoma cell and economically and non-invasively,59 and a multi­parametric
the variable number of repeats (5–11 repeats) of the MRI-based radiomics nomogram has been proposed
BamHI W region, which is the target sequence of the to provide improved prognostic ability in advanced
harmonised assay but which can vary in different viral nasopharyngeal carcinoma, suggesting that this may be a
isolates. In 2016, at a National Cancer Institute EBV promising field.60
testing harmonisation workshop for nasopharyngeal
carcinoma, nasopharyngeal carcinoma and laboratory Radiotherapy
medicine experts spoke on the limitations of the current Nasopharyngeal carcinoma is highly sensitive to ionising
quantitation assays and discussed approaches for radiation; radiotherapy is the mainstay treatment modality
improving harmonised assays in the future.50 They made for non-metastatic disease. Over time, photon-based radio­
key suggestions to guide assay harmonisation and therapy techniques have progressed from con­ ventional
validation efforts in the future; one of which was focusing two-dimensional (2D) radiotherapy to 3D conformal radio­
on an assay that would target two EBV sequences in therapy and then to intensity-modulated radiotherapy

68 www.thelancet.com Vol 394 July 6, 2019


Seminar

(IMRT). Locoregional control and survival have been Chemotherapy in non-metastatic


enhanced by the parallel improved dosimetric properties, nasopharyngeal carcinoma
and toxicity has been reduced.61–65 Currently, IMRT is While early-stage nasopharyngeal carcinoma is treated
the most widely used technique (appendix p 28). It has with only radiotherapy as the main curative treatment,
reduced the 5-year occurrence rates of locoregional failure locoregionally advanced disease requires more than
for newly diagnosed and non-metastatic naso­pharyngeal radiotherapy. Chemotherapy combined with radiotherapy
carcinoma to 7·4%.66 In a meta-analysis reviewing is a crucial development for treating locoregionally
3570 participants, IMRT was significantly associated with advanced disease. At present, the National Comprehensive
better 5-year locoregional control (odds ratio 1·94 [95% CI Cancer Network (NCCN) Guidelines recommend both
1·53–2·46]) and overall survival (1·51 [1·23–1·87]) com­ concurrent chemoradiotherapy with adjuvant chemo­
pared with 2D or 3D radiotherapy, along with signifi­cant therapy or induction chemotherapy followed by con­
reduction of radiation-induced toxicities such as temporal current chemoradiation as level 2A evidence, and
lobe neuropathy, late xerostomia, and trismus.64 concurrent chemoradiotherapy alone as level 2B evidence
While IMRT is the current preferred method, there is for stage II–IVA nasopharyngeal carcinoma.74
great interest in using proton or carbon ion radiother­apy
for treating nasopharyngeal carcinoma to improve Concurrent chemoradiotherapy
the therapeutic ratio even further. In view of the A number of trials have demonstrated the survival benefit
characteristic steep proton or carbon ion dose fall-off, of concurrent chemoradiotherapy with or without
these techniques enable a high therapeutic radiation dose adjuvant chemotherapy versus radiotherapy alone in
to be delivered to the tumour region; the exit dose is locoregionally advanced nasopharyngeal carcinoma
minimal, therefore more normal tissues are spared.67,68 (appendix pp 19–20).75–83 Recently, a meta-analysis has
Compared with IMRT, intensity-modulated proton shown that the most significant benefits of chemotherapy
therapy (IMPT) has dosimetric advantages in naso­ on overall survival are seen with either concurrent plus
pharyngeal carcinoma, with less radiation to normal adjuvant chemotherapy (HR 0·65 [95% CI 0·56–0·76]) or
structures.67 In a small study of ten patients treated with concurrent chemotherapy (0·80 [0·70–0·93]); by contrast,
IMPT with short follow-up, the radiation dose to normal there was no significant benefit following treatment with
structures was decreased, with 2-year locoregional control only adjuvant chemotherapy (0·87 [0·68–1·12]) or only
and overall survival rates of 100% and 88·9%, respec­ induction chemotherapy (0·96 [0·80–1·16]).84 However,
tively;67 further large-scaled studies with long-term whether adjuvant chemotherapy after concurrent chemo­
follow-up data are warrant. Hu and colleagues69 showed radio­therapy can confer further survival benefits remains
that intensity-modulated carbon ion therapy (IMCT) controversial. Notably, the preliminary results of a phase 3
resulted in 98·1% 1-year overall survival, 86·6% local trial85 showed no significant improvements when
recurrence–free survival, and 97·9% re­gional recurrence– adjuvant chemotherapy was added to concurrent chemo­
free survival for 75 patients with locoregionally recur­ radiotherapy versus concurrent chemoradiotherapy alone
rent nasopharyngeal carcinoma. Despite the infrequent for all outcome parameters (2-year failure-free survival,
severe toxicities reported in that study, it should be noted 86% vs 84%; overall survival, 94% vs 92%; distant
that seven patients (9·3%) developed mucosal necrosis metastasis–free survival, 88% vs 86%; locoregional
(including one fatal haemorrhage);69 future studies are failure–free survival 98% vs 95%), and the long-term
needed to provide more information on the application of results confirmed these findings (appendix p 20).86
IMCT in nasopharyngeal carcinoma. Therefore, concurrent chemoradiotherapy is deemed the
Despite the radiotherapy technique improvements, mainstay treatment in locoregionally advanced disease.
successful radiotherapy of nasopharyngeal carcinoma Concurrent chemotherapy regimens vary between
hinges on precise delineation and exact dose delivery to studies: cisplatin is commonly the first choice; dosing
the gross tumour volume (GTV), clinical target volumes schedules of 40 mg/m² once a week or 80–100 mg/m²
(CTV), and organs at risk (OARs). International every 3 weeks are acceptable in clinical practice.
consensus guidelines have been suggested for delineating Nonetheless, the differences in radiosensitivity and
the CTV and OARs to guarantee peak target coverage and toxicity profiles between different dosing schedules may
to lessen adverse effects (appendix pp 14–18),70,71 providing be neglected compared to the importance of dose
useful references for nasopharyngeal carcinoma radiation intensity, where cumulative 200 mg/m² cisplatin may be
management (appendix). Nevertheless, inter-observer the threshold for optimal efficacy without induction
variability remains high even with these contouring chemotherapy,87–89 and 160 mg/m² cisplatin when
guidelines,72 prompting efforts to improve delineation. receiving additional induction chemotherapy.90 Other
Therefore, deep learning algorithms, especially con­ alternative concurrent agents include uracil plus tegafur,
volutional neural networks, have recently been oxaliplatin, and nedaplatin (appendix pp 19–20).77,78,91
investigated for automating primary GTV delineation, In the conventional 2D radiotherapy era, Chen and
aiming to improve contouring accuracy, consistency, and colleagues92 reported significant improvements in survival
efficiency (appendix p 29).73 by concurrent chemoradiotherapy as compared to only

www.thelancet.com Vol 394 July 6, 2019 69


Seminar

radiotherapy for clinical stage II disease (appendix p 19). tegafur substantially reduced distant failure and improved
However, as IMRT has become a routine choice, many survival in high-risk patients.97,98 In an ongoing phase 3
retrospective studies have proposed that radiotherapy multicentre trial, patients with high-risk locoregionally
alone may be sufficient for treating stage II nasopharyngeal advanced nasopharyngeal carcinoma (stage III–IVA,
carcinoma.93 The results of an ongoing phase 3 clinical excluding T3–4N0 and T3N1 disease) have been randomly
trial (NCT02633202) comparing radiotherapy alone assigned to a metronomic adjuvant regimen of single-
against concurrent chemoradiotherapy in stage II and agent capecitabine (1300 mg/m² daily for 1 year) or
T3N0M0 disease might provide more evidence. observation after concurrent chemoradiotherapy with or
without induction chemotherapy (NCT02958111); the
Adjuvant chemotherapy results are highly anticipated.
Typically, adjuvant chemotherapy consists of cisplatin
(80–100 mg/m²) and fluorouracil (800–1000 mg/m² every Induction chemotherapy
day for 4–5 days) every 4 weeks over three cycles (appendix). Compared with adjuvant sequencing, induction chemo­
As mentioned above, adjuvant chemotherapy alone cannot therapy is better tolerated and eradicates micrometastases
improve survival, and additional adjuvant chemotherapy earlier; therefore, induction chemotherapy followed by
following concurrent chemoradiotherapy may not yield concurrent chemoradiotherapy may represent a prom­
further benefits in locoregionally advanced disease. A ising treatment strategy for nasopharyngeal carcinoma
phase 3 trial48 compared adjuvant chemo­therapy against in the IMRT era.99,100 However, randomised trials showed
observation for patients with high risk, in which inconsistent results regarding the efficacy of additional
104 patients identified based on detectable EBV DNA in induction chemotherapy,101–105 probably due to insufficient
the plasma after completing radiotherapy or chemo­ sample size or different induction regimens (table 2).
radiotherapy were randomly assigned to observation, or Recently, two large-scale multicentre phase 3 trials from
six-cycle adjuvant cisplatin and gemcitabine chemotherapy. Guangzhou have been reported: induction docetaxel,
This novel study represents the first biomarker-driven cisplatin, and fluorouracil106,107 significantly improved
randomised trial in nasopharyngeal carcinoma. However, 5-year overall survival (HR 0·65 [95% CI 0·43–0·98]),
despite using EBV DNA in the plasma to identify adjuvant failure-free survival (0·65 [0·43–0·98]), and distant failure-
chemotherapy patients with higher relapse risk, and using free survival (0·60 [0·38–0·95]) in locoregionally advanced
gemcitabine and cisplatin, which is more successful nasopharyngeal carcinoma (excluding N0 disease) when
than cisplatin and fluorouracil in metastatic disease,94 added to concurrent chemoradiotherapy; induction
the authors did not observe improved 5-year relapse- cisplatin and fluorouracil108 significantly improved 3-year
free survival (adjuvant chemotherapy vs obser­vation: disease-free survival (0·67 [0·47–0·95]; table 2). A small
49% vs 55%; HR 1·09 [95% CI 0·63–1·89]) or overall phase 3 trial from Tunisia and France showed that
survival (64% vs 68%; 1·09 [0·56–2·11]). The ongoing induction docetaxel, cisplatin, and fluorouracil signifi­
NRG-HN001 trial (NCT02135042) also uses post- cantly improved 3-year progression-free survival (HR 0·44
radiotherapy plasma EBV DNA to establish whether [95% CI 0·20–0·97]), with a marginally significant effect
adjuvant gemcitabine and paclitaxel is better than cisplatin on overall survival (0·40 [0·15–1·04]),109 confirming the
and fluorouracil for patients with detectable EBV DNA in efficacy of this triplet regimen. Another phase 3 trial from
the plasma, and whether adjuvant cisplatin and fluorouracil Taiwan observed improved 5-year disease-free survival
can be omitted in patients with undetectable plasma EBV (HR 0·74 [95% CI 0·57–0·97]) by adding induction
DNA; it might provide more evidence for biomarker- mitomycin, epirubicin, cisplatin, fluorouracil, and
guided use of conventional adjuvant chemotherapy. leucovorin, although overall survival was not improved.110
One potential reason for the poor efficacy of adjuvant Considering the controversial results, pooled analysis of
chemotherapy may be the poor tolerance of conventional individual patient information from four randomised
chemotherapy regimens (cisplatin and fluorouracil/ trials from endemic areas was performed: induction
gemcitabine) in the first 6 months after radiotherapy chemotherapy additional to concurrent chemoradiotherapy
completion, with 50–76% compliance at best.79–83,85,94 significantly improved overall survival (HR 0·75 [95% CI
Metronomic use of oral chemotherapy agents such as 0·57–0·99]; 6% absolute benefit at 5 years) with the
capecitabine or uracil plus tegafur as adjuvant chemo­ survival benefit mainly associated with improved distant
therapy may be an alternative choice after concurrent control (0·68 [95% CI 0·51–0·90]; 7% absolute reduction
chemoradiotherapy. Metronomic chemotherapy describes in distant failure rate at 5 years).111 To summarise eisting
the close, regular administration of chemotherapy drugs at studies, the 2018 NCCN Guidelines upgraded the evidence
less toxic doses over prolonged periods, with the advantages for induction chemotherapy plus concurrent chemo­
of good compliance, low toxicities, and convenience.95 The radiotherapy from level 3 to level 2A, which is equal to that
mechanisms of metronomic chemotherapy include anti- of concurrent chemoradiotherapy plus adjuvant chemo­
angiogenic effects, induction of tumour dormancy, and therapy.74 In summary, induction chemotherapy plays an
activation of immunity.95,96 Two retrospective analyses increasingly important role in managing locoregionally
showed that 12-month metronomic adjuvant uracil plus advanced nasopharyngeal carcinoma in the IMRT era,

70 www.thelancet.com Vol 394 July 6, 2019


Seminar

Experimental chemotherapy Control chemotherapy Inclusion Sample Overall survival Progression-free survival
period size
Experimental vs HR (95% CI); Experimental vs HR (95% CI);
control (n-year p value control (n-year p value
results) results)
Induction chemotherapy plus concurrent chemoradiotherapy vs concurrent chemoradiotherapy alone
Hui et al101 Induction: docetaxel 75 mg/m² d1; cisplatin Concurrent: cisplatin 2002–04 68 94% vs 68% (3) 0·24 88% vs 60% (3) 0·49
(Phase 2) 75 mg/m² d1; q3wks × 2; concurrent: cisplatin 40 mg/m² d1; q1wk × 8 (0·08–0·73); (0·20–1·19);
40 mg/m² d1; q1wk × 8 p=0·012 p=0·12
Fountzilas et al102 Induction: cisplatin 75 mg/m² d1; epirubicin Concurrent: cisplatin 2003–08 141 70% vs 59% (3) 0·95 65% vs 64% (3) 1·40
(Phase 2) 75 mg/m2 d1; paclitaxel 175 mg/m² d1; 40 mg/m2 d1; q1wk × 8 (0·48–1·89); (0·71–2·77);
q3wks × 3; concurrent: cisplatin 40 mg/m² p=0·89 p=0·38
d1; q1wk × 8
Tan et al103 Induction: gemcitabine 1000 mg/m² d1, d8; Concurrent: cisplatin 2004–12 172 94% vs 92% (3) 1·05 (0–2·19); 75% vs 67% (3) 0·77
(Phase 3) carboplatin AUC=2·5 d1, d8; paclitaxel 40 mg/m² d1; q1wk × 8 p=0·49 (0·44–1·35);
70 mg/m² d1, d8; q3wks × 3; concurrent: p=0·36
cisplatin 40 mg/m² d1; q1wk × 8
Sun et al106,107 Induction: docetaxel 60 mg/m² d1; cisplatin Concurrent: cisplatin 2011–13 480 86% vs 78% (5) 0·65 77% vs 66% (5) 0·65
(Phase 3) 60 mg/m² d1; fluorouracil 600 mg/m² d1-5; 100 mg/m² d1; q3wks × 3 (0·43–0·98); (0·43–0·98);
q3wks × 3; concurrent: cisplatin 100 mg/m² p=0·042 p=0·019
d1; q3wks × 3
Cao et al108 Induction: cisplatin 80 mg/m² d1; Concurrent: cisplatin 2008–15 476 88% vs 89% (3) NR; p=0·82 82% vs 74% (3) 0·67
(Phase 3) fluorouracil 800 mg/m² d1-5; q3wks × 2; 80 mg/m2 d1; q3wks × 3 (0·47–0·95);
concurrent: cisplatin 80 mg/m² d1; q3wks × 3 p=0·028
Frikha et al109 Induction: docetaxel 75 mg/m² d1; cisplatin Concurrent: cisplatin 2009–12 83 86% vs 69% (3) 0·40 74% vs 57% (3) 0·44
(Phase 3) 75 mg/m² d1; fluorouracil 750 mg/m² d1-5; 40 mg/m² d1; q1wk × 7 (0·15–1·04); (0·20–0·97);
q3wks × 3; concurrent: cisplatin 40 mg/m² p=0·059 p=0·042
d1; q1wk × 7
Hong et al110 Induction: mitomycin 8 mg/m² d1; Concurrent: cisplatin 2003–09 479 72% vs 68% (5) 0·92 61% vs 50% (5) 0·74
(Phase 3) epirubicin 60 mg/m² d1; cisplatin 60 mg/m² 30 mg/m² d1; q1wk (0·67–1·27); (0·57–0·97);
d1; fluorouracil 450 mg/m² d8; leucovorin p=0·62 p=0·026
30 mg/m² d8; concurrent: cisplatin
30 mg/m² d1; q1wk
Others
Huang et al104 Induction: fluorouracil 750 mg/m² d1-5; Induction: fluorouracil 750 2002–05 400 50% vs 49% (10) 0·95 48% vs 48% (10) 1·01
(Phase 3) carboplatin AUC=6 d1; q3wks × 2; mg/m² d1-5; carboplatin (0·72–1·26); (0·77–1·33);
concurrent: carboplatin AUC=6 d7; q3wks × 3 AUC=6 d1; q3wks × 2 p=0·71 p=0·94
Lee et al105 Induction: cisplatin 100 mg/m² d1; Concurrent: cisplatin 2006–12 390 88% vs 83% (3) NR; p=0·12 80% vs 75% (3) NR; p=0·073
(Phase 3) fluorouracil 1000 mg/m² d1-5; or cisplatin 100 mg/m² d1; q3wks × 2/3; (706)*
100 mg/m² d1; capecitabine 2000 mg/m² adjuvant: cisplatin
d1-14; q3wks × 3; concurrent: cisplatin 80 mg/m² d1; fluorouracil
100 mg/m² d1; q3wks × 2/3 1000 mg/m² d1-4,
q4wks × 3
(Table 2 continues on next page)

which helps further improve distant control and sub­ greatly anticipated. Replacing cisplatin with lobaplatin
sequently survival; patients with high risk of distant or nedaplatin, or fluorouracil with capecitabine during
metastasis may benefit from additional induction chemo­ induction and concurrent phases may help maintain non-
therapy over concurrent chemoradiotherapy alone. inferior efficacy with reduced toxicities and improved
However, the ideal intensity for induction chemo­ ­ quality of life and convenience (ChiCTR-TRC-13003285,
therapy has not been established. The high-intensity NCT03503136; table 2). Furthermore, effective biomark­ers
docetaxel plus cisplatin plus fluorouracil regimen appears predicting ideal candidates for induction chemo­therapy
to confer more benefits than the docetaxel plus cisplatin remain investigational. Pre-treatment EBV DNA or
or cisplatin plus fluorouracil combinations,101,106–108 but relevant genomic signatures may help identify patients
statis­
tically significant survival differences between most likely to benefit from induction chemotherapy; EBV
induction regimens were not detected by pooled analysis.111 DNA surveillance during induction chemotherapy may
Future prospective trials should be performed to identify also provide real-time information on tumour response
the ideal induction chemotherapy regimens and cycles. for therapeutic adaptation.
The gemcitabine and cisplatin regimen has demon­
strated its superiority in metastatic nasopharyngeal Disease surveillance and toxic effects
carcinoma,94 and is being tested as an induction regimen Local, regional, and/or distant failure can occur in non-
in a phase 3 trial (NCT01872962), with the results being metastatic disease despite aggressive treatment. High-dose

www.thelancet.com Vol 394 July 6, 2019 71


Seminar

Experimental chemotherapy Control chemotherapy Inclusion Sample Overall survival Progression-free survival
period size
Experimental vs HR (95% CI); Experimental vs HR (95% CI);
control (n-year p value control (n-year p value
results) results)
(Continued from previous page)
Phase 3 trials in progress
Ma et al Induction: gemcitabine 1000 mg/m² d1, d8; Concurrent: cisplatin 2013–16 480 ·· ·· ·· ··
(NCT01872962) cisplatin 80 mg/m² d1; q3wks × 3; 100 mg/m2 d1; q3wks × 3
concurrent: cisplatin 100 mg/m² d1;
q3wks × 3
Guo et al (ChiCTR- Induction: lobaplatin 30 mg/m2 d1; Induction: cisplatin 2013–16 494 ·· ·· ·· ··
TRC­13003285) fluorouracil 800 mg/m2 d1–5; q3wks × 2; 100 mg/m² d1; fluorouracil
concurrent: lobaplatin 30 mg/m2 d1; 800 mg/m² d1-5;
q3wks × 2 q3wks × 2; Concurrent:
cisplatin 100 mg/m² d1;
q3wks × 2
Mai et al Induction: paclitaxel liposome 135 mg/m2 Concurrent: cisplatin 2017– 322 ·· ·· ·· ··
(NCT03306121) d1; cisplatin 25 mg/m2 d1–3; fluorouracil 100 mg/m² d1; q3wks × 3; ongoing
750 mg/m2 d1–5; q3wks × 3; concurrent: Adjuvant: cisplatin
cisplatin 100 mg/m2 d1; q3wks × 3 80 mg/m² d1; fluorouracil
1000 mg/m² d1–4,
q4wks × 3
Ma et al Group A: Induction: docetaxel 60 mg/m² d1; ·· 2018– 632 ·· ·· ·· ··
(NCT03503136) cisplatin 60 mg/m² d1; fluorouracil ongoing
600 mg/m² d1–5; q3wks × 3; concurrent:
cisplatin 100 mg/m² d1; q3wks × 2; Group B:
Induction: docetaxel 60 mg/m² d1;
nedaplatin 60 mg/m² d1; fluorouracil
600 mg/m² d1–5; q3wks × 3; concurrent:
nedaplatin 100 mg/m² d1; q3wks × 2;
Group C: Induction: docetaxel 60 mg/m² d1;
cisplatin 60 mg/m² d1; capecitabine
1250 mg/m2 d1–14; q3wks × 3; concurrent:
cisplatin 100 mg/m² d1; q3wks × 2; Group D:
Induction: docetaxel 60 mg/m2 d1;
nedaplatin 60 mg/m² d1; capecitabine
1250 mg/m² d1–14; q3wks × 3; concurrent:
nedaplatin 100 mg/m² d1; q3wks × 2
AUC=area under the concentration–time curve. ChiCTR=Chinese Clinical Trial Register. HR=hazard ratio. NCT=ClinicalTrials.gov identifier. NR=not reported. q1/3/4wks=every 1/3/4 weeks. *390 patients in
conventional fractionation radiotherapy arms; 706 patients in the whole trial.

Table 2: Randomised trials evaluating induction chemotherapy plus concurrent chemoradiotherapy

radiation or chemoradiotherapy inevitably induces acute value to the diagnosis, as it is more accurate in
and late toxicities, and the latter can emerge months or distinguishing residual or recurrent disease from fibrosis
even years after treatment completion. These issues or scar tissue post radiotherapy, and combined MR and
highlight the importance of close follow-up. ¹⁸F-FDG-PET/CT is more accurate for tumour restaging
12 weeks after the completion of radiotherapy or than either modality independently.55,114 Plasma EBV DNA
chemoradiotherapy is widely considered the appropriate could also be used as a complementary surveillance
timepoint for initial assessment of residual disease, as by method to conventional imaging for monitoring failure,
then, treatment-induced inflammation would have largely particularly distant metastasis.115,116 A prospective, multi­
resolved, most tumours would have regressed, and delayed centre trial has confirmed that post-radiotherapy plasma
tumour regression within this timepoint (12 weeks) EBV DNA levels correlate significantly with the
would not affect overall local control.112,113 Comprehensive corresponding hazards of locoregional failure, distant
response assessment includes thorough history and metastasis, and death in nasopharyngeal carcinoma
physical examination, nasopharyngoscopy with or without (appendix p 21).48
biopsy, plasma EBV DNA, and radiological imaging. It The most commonly observed acute radiotherapy-
represents a challenge for distinguishing residual tumours related toxicities in IMRT for nasopharyngeal carcinoma
from post radiation changes. Although MRI is superior to include mucositis, dermatitis, xerostomia, and dysphagia
CT for diagnosing local residual/recurrent nasopharyn­ (appendix p 30). Adding chemotherapy to radiotherapy
geal carcinoma, both modalities have unsatisfactorily low invariably increases haematological and non-haemato­
accuracy.111,112 Here, ¹⁸F-FDG-PET/CT can add additional logical acute toxicity incidence. Although pharmacological

72 www.thelancet.com Vol 394 July 6, 2019


Seminar

interventions for acute mucositis and xerostomia are often Management of metastatic disease
ineffective, clinical symptoms generally improve within Patients with metastatic nasopharyngeal carcinoma (stage
weeks after treatment cessation. Sometimes, grade 3 or IVB) are heterogeneous groups with different outcomes;
4 reactions can persist and lead to consequential late in some cases, long-term survivorship is possible. For
effects. Surveillance and prompt management of late example, in synchronous metastases, which are present in
toxicities are important for survivors’ long-term wellbeing. approximately 10% of newly diagnosed patients, prognosis
Common late toxicities include xerostomia, sensorineural is closely related with the anatomic degree of metastasis
hearing loss, osteoradionecrosis, trismus, CNS abnor­ lesions; palliative chemotherapy can lead to median
malities (eg, temporal lobe injury), and hormonal dys­ overall survival of approximately 10–15 months, which can
function (eg, hypothyroidism; appendix p 30). be greatly increased if the patient is suitable for com­bined
therapy such as locoregional radiotherapy and local
Managing residual or recurrent disease therapy of metastatic lesions.127–131 Therefore, individualised
In the IMRT era, around 10% of patients have residual treatment for metastatic nasopharyngeal carcinoma is
disease or develop recurrent disease at the primary necessary. The risk stratification for metastatic naso­
and/or regional site.66,117 It is widely acknowledged that pharyngeal carcinoma can be performed based on clinical
neck dissection is the primary choice for patients with features such as metastasis status (e.g. oligo­metastases,
isolated regional failure; the 5-year overall survival rate metachronous metastasis, lung or liver solitary metas­
is 41%.118 Radiotherapy or surgery can salvage local tasis, multiple liver metastases), or the incorporation of
nasopharyngeal failure. Commonly, tumours that recur prognostic markers (eg—haemoglobin, lactate dehy­
within 1 year are considered radioresistant; surgery is drogenase, C-reactive protein or albumin).130,132–136
recommended if they are resectable. Recently, a In prognostic groups with such favourable features, a
prognostic model119 was developed for stratifying curative tactic that involves the primary tumour receiving
radioresistant nasopharyngeal carcinoma, where low- radical chemoradiotherapy and oligometastases under­
risk patients are ideal candidates for curative repeat going ablative treatment may be optimal. In terms of
IMRT. In addition, as previously mentioned, IMCT systemic treatment, the platinum-containing doublet of
could be a promising salvage treatment for recurrent cisplatin plus fluorouracil has been the first-line therapy
nasopharyngeal carcinoma, considering its physical/ for recurring or metastatic disease for decades,
biological advantages over IMRT.69 Nonetheless, repeat with 40–65% response rates.137–139 Nonetheless, whether
irradiation should be approached with caution in all cisplatin plus fluorouracil represents the most efficacious
cases, as there is a high risk of fatal late effects, with treatment strategy has always been contentious, as it has
a recent meta-analysis reporting 33% grade 5 toxicity never been directly compared with other regimens in
following repeat IMRT.120 In summary, aggressive treat­ randomised trials.140 In the landmark GEM20110714 trial,141
ment including surgery or re-irradiation may be better Zhang and colleagues presented for the first time a
for improving the chances of long-term survival in the randomised phase 3 trial of two competing combinations
management of recurrent nasopharyngeal carcinoma, containing platinum: gemcitabine  plus cisplatin versus
while chemotherapy may be better for residual/ fluorouracil plus cisplatin, in patients with recurrent or
recurrent disease that is neither resectable nor suitable metastatic disease. Inspiringly, a significantly higher
for re-irradiation, or for high-risk patients with poor proportion of patients in the gemcitabine group achieved
prognosis.121 an objective response than in the fluorouracil group
Given the deep location and complex adjacency of the (64% vs 42%; p<0·0001). Compared with fluorouracil plus
nasopharynx, salvage surgery of local failure can be cisplatin, gemcitabine plus cisplatin also significantly
challenging, with potentially severe morbidities. Although improved progression-free survival (median: 7·0 vs
various open approaches such as the transmaxillary 5·6 months; HR 0·55 [95% CI 0·44–0·68]) and overall
approach have been devised for nasopharyngectomy,122 survival (median: 29·1 vs 20·9 months; 0·62 [0·45–0·84]).
where the 5-year disease-free survival rate can reach 57%, That study establishes gemcitabine plus cisplatin as a
>40% of patients experience complications such as facial new standard first-line treatment option for recurrent or
numbness, trismus, and palatal fistula.123 To avoid the metastatic nasopharyngeal carcinoma.
grave complications of open surgery, Chen and colleagues The progress of molecular-targeted therapies in
developed an endoscopic nasopharyngectomy technique nasopharyngeal carcinoma is lagging (appendix), and
that allowed en-bloc tumour resection similar to open more large-scale randomised studies are warranted
surgery; they further adapted a modified pedicle nasal to assess the value of targeted drugs in nasopharyn­
septum and floor mucosa flap to resurface the geal carcinoma. Encouragingly, immune checkpoint
nasopharyngeal defect that is very difficult to heal due to therapies have achieved great progress in recurrent or
the prior radiation.124,125 A new surgical staging system of metastatic nasopharyngeal carcinoma, and represent
recurrent nasopharyngeal carcinoma has also been a promising approach towards improving clinical
proposed to help determine whether the disease is outcomes, which will be discussed in the following
resectable.126 section.

www.thelancet.com Vol 394 July 6, 2019 73


Seminar

Immunotherapy for nasopharyngeal carcinoma checkpoint blockades. EBV-directed vaccination exploits


Tumour immunotherapy may represent a promising T-cell activation to target nasopharyngeal cancer cells by
therapeutic approach in nasopharyngeal carcinoma, recognising expressed viral antigens (eg—EBNA1, LMP1,
where the primary strategies include EBV-directed LMP2). For example, vaccination against LMP2 epitopes
vaccination, adoptive T-cell therapy, and immune with dendritic cells or peptides, or adoptive transfer of

Key eligibility criteria Experimental regimen Control regimen Sample Objective Overall survival Progression-free
size response survival
Median 1-year Median 1-year
(months) rate (months) rate
Trials with results
Hsu et al142 Recurrent or metastatic disease; Pembrolizumab 10 mg/kg every ·· 27 26% 16·5 63% 6·5 33%
(phase 1) failure on previous standard 2 weeks up to 2 years or until
therapy; PD-L1 expression in 1% disease progression or unacceptable
or more of tumour cells or toxicity
tumour-infiltrating lymphocytes
Ma et al143 Recurrent or metastatic disease; Nivolumab 3 mg/kg every 2 weeks ·· 44 20% 17·1 59% 2·8 19%
(phase 2) failure on at least one previous on a 4-week cycle until disease
line of platinum-based progression
chemotherapy
Fang et al144 Recurrent or metastatic disease; Camrelizumab at escalating doses ·· 93 34% NR NR 5·6 27%
(phase 1) failure on at least one previous of 1 mg/kg, 3 mg/kg, and 10 mg/kg,
line of platinum-based and a bridging dose of 200 mg
chemotherapy per dose once every 2 weeks until
unacceptable toxicity
Fang et al144 Recurrent or metastatic disease; Camrelizumab 200 mg (day 1), ·· 23 91% NR NR NR 61%
(phase 1) treatment-naive patient gemcitabine 1 g/m² (days 1 and 8),
and cisplatin 80 mg/m² (day 1)
every 3 weeks for six cycles,
followed by camrelizumab 200 mg
maintenance once every 3 weeks
Phase 3 trials in progress
Merck Sharp & Recurrent or metastatic disease; Pembrolizumab 200 mg every Capecitabine 2000 mg/m² 230 ·· ·· ·· ·· ··
Dohme Corp failure on at least one previous 3 weeks on a 3-week cycle until d1-14 of each 3-week cycle or
(NCT02611960) line of platinum-based disease progression or unacceptable gemcitabine 1250 mg/m² d1,
chemotherapy toxicity or a maximum of up to d8; of each 3-week cycle or
35 cycles docetaxel 75 mg/m² d1 of each
3-week cycle until disease
progression or unacceptable
toxicity
Xu et al Recurrent or metastatic disease; JS001 combined with gemcitabine Placebo combined with 280 ·· ·· ·· ·· ··
(NCT03581786) treatment-naive patient and cisplatin given every 3 weeks in gemcitabine and cisplatin
3-week cycles given every 3 weeks in 3-week
cycles
Zhang et al Recurrent or metastatic disease; Camrelizumab 200 mg (day 1), Placebo combined with 250 ·· ·· ·· ·· ··
(NCT03707509) treatment-naive patient gemcitabine 1 g/m² (days 1 and 8), gemcitabine 1 g/m² (days 1
and cisplatin 80 mg/m² (day 1) and 8), and cisplatin 80 mg/m²
every 3 weeks for six cycles (day 1) every 3 weeks for
six cycles
Ma et al Stage III-IVA disease (except Adjuvant: camrelizumab 3 mg/kg Observation 400 ·· ·· ·· ·· ··
(NCT03427827) T3-4N0 and T3N1); completed (≤200 mg) d1; q4wks × 12
induction chemotherapy of
gemcitabine and cisplatin
followed by concurrent
cisplatin-radiotherapy;
4-6 weeks after chemoradiation
Ma et al Stage III-IVA disease (except Induction: gemcitabine Induction: gemcitabine 420 ·· ·· ·· ·· ··
(NCT03700476) T3–4N0 and T3N1) 1000 mg/m² d1, d8; cisplatin 1000 mg/m² d1, d8; cisplatin
80 mg/m² d1; sintilimab 200 mg 80 mg/m² d1; q3wks × 3;
d1; q3wks × 3; concurrent: cisplatin Concurrent: cisplatin
100 mg/m² d1; q3wks × 2; sintilimab 100 mg/m² d1; q3wks × 2
200 mg d1; q3wks × 3
NCT=ClinicalTrials.gov identifier. NR=not reported. PD-L1=programmed death-ligand 1. q3/4wks=every 3/4 weeks.

Table 3: Clinical trials evaluating immune checkpoint inhibitors in nasopharyngeal carcinoma

74 www.thelancet.com Vol 394 July 6, 2019


Seminar

cytotoxic T lymphocytes or TILs have shown potential


clinical efficacy in advanced nasopharyngeal carcinoma Panel: Research questions on nasopharyngeal carcinoma that remain to be answered
(appendix). • What biological mechanisms underlie the progress of Epsten-Barr virus (EBV)
Recently, immune checkpoint blockade therapies have infection and the development of nasopharyngeal carcinoma?
achieved breakthroughs in tumour immnuotherapy. • What are the progressive genomic changes during nasopharyngeal carcinoma
Nasopharyngeal carcinoma tumours are characterised development, progression, and recurrence?
by high PD-L1 expression (up to 90% of tumour cells) • Which population should be screened for nasopharyngeal carcinoma, and how and at
and abundant infiltration of non-malignant lymphocytes what age and interval should this be done?
(about 50% of samples with >70% stromal TILs or • How can plasma EBV DNA be incorporated into the current staging system?
>10% intratumoral TILs; appendix),37–42 which renders • How can longitudinal plasma EBV DNA surveillance during treatment be applied for
patients potentially suitable for immune checkpoint risk-stratified therapeutic adaptation?
blockade therapies. Several important single-arm trials • How can biomarkers for predicting treatment response (eg, induction chemotherapy,
evaluating anti–PD-1 monoclonal antibodies in recur­ adjuvant chemotherapy, immunotherapy) and prognosis be developed and
rent or metastatic nasopharyngeal carcinoma have been validated?
reported, where PD-1 inhibitors showed promising • How can reliable radiomics models for improving decision support in nasopharyngeal
clinical activity (table 3).142–144 In a phase 1 trial of 27 pa­­ carcinoma be developed?
tients with unresectable or metastatic nasopharyngeal • How can artificial intelligence automation for nasopharyngeal carcinoma treatment
carcinoma that had failed prior standard therapy and had decision (radiotherapy planning, chemotherapy timing and regimens) be applied?
PD-L1 expression in ≥1% of tumour cells or TILs, • What is the emerging role of IMPT and IMCT in nasopharyngeal carcinoma?
pembrolizumab treatment resulted in 26% objective • What is the best possible way to administer concurrent cisplatin, tri-weekly or
response, 63% 1-year overall survival, and 33% 1-year weekly?
progression-free sur­vival.142 In another phase 2 trial • Should stage II or T3N0M0 nasopharyngeal carcinoma be managed by radiotherapy
evaluating nivolumab in 44 patients with multiply pre- alone or concurrent chemoradiotherapy?
treated recurrent or metastatic nasopharyngeal carci­ • Could lobaplatin or nedaplatin replace cisplatin, or could capecitabine replace
noma, the objective response rate was 20%, with fluorouracil?
59% 1-year overall survival and 19% 1-year progression- • What is the value of metronomic adjuvant chemotherapy (eg, low-dose oral
free survival.143 Recently, Fang and colleagues144 reported fluorouracil such as capecitabine) in locoregionally advanced nasopharyngeal
on two phase 1 trials, where camrelizumab (anti–PD-1 carcinoma?
monoclonal antibody) was used as (1) monotherapy • Which patients with residual or recurrent nasopharyngeal carcinoma should receive
for 93 patients with multiply pre-treated recurrent surgery or radiotherapy?
or metastatic disease, and (2) in combination with • Which patients with metastatic nasopharyngeal carcinoma should receive definitive
gemcitabine plus  cisplatin for 23 patients with treatment- radiotherapy or chemoradiotherapy?
naïve recurrent or metastatic disease. In the monother­ • What is the role of metastasectomy or stereotactic ablative radiotherapy in
apy trial, there was 34% objective response rate and oligometastatic nasopharyngeal carcinoma?
27% 1-year progression-free survival. In the combination • How can immunotherapy, especially immune checkpoint blockade, be incorporated
trial, the objective response rate was 91% and the into the current treatment paradigm of nasopharyngeal carcinoma?
1-year progression-free survival rate was 61%. As • How can patients that could benefit from immunotherapy be identified?
anti-PD-1 antibodies have promising anti-tumour activity
and predictable safety profile, several randomised
phase 3 trials for nasopharyngeal carcinoma are ongoing One future challenge is that PD-1 inhibition benefits
(table 3). For recurrent or metastatic disease, the only a subset of patients. Identifying biomarkers
KEYNOTE-122 trial (NCT02611960) aims to evaluate associated with treatment response would allow appro­
pembrolizumab monotherapy against standard chemo­ priate immunotherapy tailoring for different patient
therapy in previously treated patients, while another subgroups. Unfortunately, no strong evidence currently
two trials (NCT03581786 and NCT03707509) are further supports the correlation between response rate to anti-
assessing anti–PD-1 antibody combined with gemcitabine PD-1 monoclonal antibodies and known biomarkers such
and cisplatin versus gemcitabine and cisplatin for as PD-L1 expression in tumour or immune cells, HLA-A
treatment-naive patients. These studies’ final results will and HLA-B expression, pre-treatment or post-treatment
show whether anti–PD-1 antibody is preferred for EBV DNA levels, plasma EBV DNA clearance, and
salvaging chemotherapy failure or whether the combined tumour mutational burden in advanced nasopharyngeal
method is more suitable as a first-line standard of care for carcinoma.143,144 Nonetheless, these exploratory biomarker
appro­priate patients. Two phase 3 trials (NCT03427827 analyses were insufficiently powered, and future large-
and NCT03700476) investigating adjuvant camrelizumab scale studies are warranted to identify and validate
or induction and concurrent sintilimab (anti-PD-1 potential therapeutic biomarkers (eg, TIL level, immune
antibody) in locoregionally advanced nasopharyngeal gene signatures, immune molecular subgroups)145–151
carcinoma will test the value of adding anti-PD-1 therapy associated with nasopharyngeal carcinoma treatment
to standard treatment in the curative setting. response or resistance.

www.thelancet.com Vol 394 July 6, 2019 75


Seminar

Apart from biomarker identification, a bigger patient Sun Yat-sen University Cancer Center, Guangzhou, China) for assistance
group might require an immuno-oncology cocktail. with providing the figure materials.
Fang and colleagues144 reported on camrelizumab mono­ References
1 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A.
therapy in eight patients who had received ipilimumab Global cancer statistics 2018: GLOBOCAN estimates of incidence
previously; they had a significantly higher response to and mortality worldwide for 36 cancers in 185 countries.
the consequent camrelizumab compared to patients CA Cancer J Clin 2018; 68: 394–424.
without prior ipilimumab (75% vs 30%). These results 2 Ferlay J, Ervik M, Lam F, et al. Global Cancer Observatory:
cancer today. Lyon, France: International Agency for Research on
suggest that dual immune checkpoint inhibition, such as Cancer 2018. https://gco.iarc.fr/today (accessed Dec 28, 2018).
combination or sequential therapy with antibodies 3 Tang LL, Chen WQ, Xue WQ, et al. Global trends in incidence and
against PD-1, CTLA-4, and other immune checkpoint mortality of nasopharyngeal carcinoma. Cancer Lett 2016; 374: 22–30.
4 Wei KR, Zheng RS, Zhang SW, Liang ZH, Li ZM, Chen WQ.
agents may further improve clinical activity. Nasopharyngeal carcinoma incidence and mortality in China, 2013.
Chin J Cancer 2017; 36: 90.
Conclusions and future directions 5 Lee AW, Foo W, Mang O, et al. Changing epidemiology of
nasopharyngeal carcinoma in Hong Kong over a 20-year period
Nasopharyngeal carcinoma incidence has declined (1980–99): an encouraging reduction in both incidence and
gradually but progressively, accompanied by substantially mortality. Int J Cancer 2003; 103: 680–5.
reduced mortality, which is likely due to lifestyle and 6 Li K, Lin GZ, Shen JC, Zhou Q. Time trends of nasopharyngeal
carcinoma in urban Guangzhou over a 12-year period (2000–2011):
environmental changes, enhanced understanding of the declines in both incidence and mortality. Asian Pac J Cancer Prev
pathogenesis and risk factors, population screening, 2014; 15: 9899–903.
advancements in diagnostic techniques, and individualised 7 Chen W, Zheng R, Baade PD, et al. Cancer statistics in China, 2015.
comprehensive chemoradiotherapy strategies. However, CA Cancer J Clin 2016; 66: 115–32.
8 Yu WM, Hussain SS. Incidence of nasopharyngeal carcinoma in
considering the current lack of reliable evidence, several Chinese immigrants, compared with Chinese in China and
questions on nasopharyngeal carcinoma pathogenesis and South East Asia: review. J Laryngol Otol 2009; 123: 1067–74.
clinical management remain to be addressed (panel). The 9 Wang HY, Chang YL, To KF, et al. A new prognostic histopathologic
classification of nasopharyngeal carcinoma. Chin J Cancer 2016;
next decade of nasopharyngeal carcinoma research will 35: 41.
focus on exploring the biological mechanisms underlying 10 Pathmanathan R, Prasad U, Chandrika G, Sadler R, Flynn K,
its pathogenesis, refining screening and staging strategies, Raab-Traub N. Undifferentiated, nonkeratinizing, and squamous
cell carcinoma of the nasopharynx. Variants of Epstein-Barr
identifying biomarkers related to prognostic risk strati­ virus-infected neoplasia. Am J Pathol 1995; 146: 1355–67.
fication and treatment benefits gained, optimising 11 Young LS, Dawson CW. Epstein-Barr virus and nasopharyngeal
treatment strategies for the different patient subgroups, carcinoma. Chin J Cancer 2014; 33: 581–90.
and developing novel therapeutics. Hopefully, ongoing 12 Chua MLK, Wee JTS, Hui EP, Chan ATC. Nasopharyngeal
carcinoma. Lancet 2016; 387: 1012–24.
studies will identify the needed knowledge, tools, and 13 Bei JX, Zuo XY, Liu WS, Guo YM, Zeng YX. Genetic susceptibility
therapies for advancing the management of naso­ to the endemic form of NPC. Chin Clin Oncol 2016; 5: 15.
pharyngeal carcinoma to the next level. 14 Bei JX, Li Y, Jia WH, et al. A genome-wide association study of
nasopharyngeal carcinoma identifies three new susceptibility loci.
Contributors Nat Genet 2010; 42: 599–603.
YPC, YS, and JM were involved in the conception of the manuscript. 15 Fachiroh J, Sangrajrang S, Johansson M, et al. Tobacco consumption
YPC, ATCC, QTL, PB, YS, and JM wrote and reviewed the original and genetic susceptibility to nasopharyngeal carcinoma (NPC) in
manuscript. YPC, ATCC, QTL, PB, YS, and JM revised the manuscript. Thailand. Cancer Causes Control 2012; 23: 1995–2002.
YPC, ATCC, QTL, PB, YS, and JM approved the final manuscript. 16 Ng CC, Yew PY, Puah SM, et al. A genome-wide association study
Declaration of interests identifies ITGA9 conferring risk of nasopharyngeal carcinoma.
ATCC has received clinical research funding from Bristol-Myers Squibb, J Hum Genet 2009; 54: 392–97.
Boehringer Ingelheim, and Pfizer Oncology outside the submitted work. 17 Bei JX, Su WH, Ng CC, et al. A GWAS meta-analysis and replication
PB has received funding from Astellas Pharma, Merck-Serono, study identifies a novel locus within CLPTM1L/TERT associated
with nasopharyngeal carcinoma in individuals of Chinese ancestry.
and Amgen outside the submitted work. All other authors declare no
Cancer Epidemiol Biomarkers Prev 2016; 25: 188–92.
competing interests.
18 Cui Q, Feng QS, Mo HY, et al. An extended genome-wide
Acknowledgments association study identifies novel susceptibility loci for
We would like to thank Melvin L K Chua (Division of Radiation Oncology, nasopharyngeal carcinoma. Hum Mol Genet 2016; 25: 3626–34.
National Cancer Centre Singapore; Oncology Academic Clinical 19 Dai W, Zheng H, Cheung AK, et al. Whole-exome sequencing
Programme, Duke-NUS Medical School, Singapore) for his helpful identifies MST1R as a genetic susceptibility gene in
comments on this study. We thank Ming-Yuan Chen (Department of nasopharyngeal carcinoma. Proc Natl Acad Sci USA 2016;
Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 113: 3317–22.
Guangzhou, China) for assisting with the writing of Management of 20 Lin DC, Meng X, Hazawa M, et al. The genomic landscape of
Residual or Recurrent Disease section. We thank Kwok-Wai Lo nasopharyngeal carcinoma. Nat Genet 2014; 46: 866–71.
(Department of Anatomical & Cellular Pathology, The Chinese University 21 Zheng H, Dai W, Cheung AK, et al. Whole-exome sequencing
of Hong Kong, Hong Kong, China) for providing materials and identifies multiple loss-of-function mutations of NF-kappaB
pathway regulators in nasopharyngeal carcinoma.
comments on the plotting of figure 2. We thank Mu-Sheng Zeng,
Proc Natl Acad Sci USA 2016; 113: 11283–88.
Wei-Hua Jia, and Jin-Xin Bei (State Key Laboratory of Oncology in
22 Li YY, Chung GT, Lui VW, et al. Exome and genome sequencing of
South China, Sun Yat-sen University Cancer Center, Guangzhou, China)
nasopharynx cancer identifies NF-kappaB pathway activating
for their comments on the Pathology and Risk Factors section. We thank mutations. Nature Commun 2017; 8: 14121.
Zi-Qi Zheng and Wei-Jie Luo (Department of Radiation Oncology,
23 Zhang L, MacIsaac KD, Zhou T, et al. Genomic analysis of
Sun Yat-sen University Cancer Center, Guangzhou, China) for assistance nasopharyngeal carcinoma reveals TIME-based subtypes.
with figure plotting. We thank Ling-Long Tang, Li Lin, Jia-Wei Lv, Mol Cancer Res 2017; 15: 1722–32.
Yuan Zhang, and Ya-Qin Wang (Department of Radiation Oncology,

76 www.thelancet.com Vol 394 July 6, 2019


Seminar

24 Guo X, Johnson RC, Deng H, et al. Evaluation of nonviral risk 45 Guo R, Tang LL, Mao YP, et al. Proposed modifications and
factors for nasopharyngeal carcinoma in a high-risk population of incorporation of plasma Epstein-Barr virus DNA improve the
Southern China. Int J Cancer 2009; 124: 2942–47. TNM staging system for Epstein-Barr virus-related nasopharyngeal
25 Tsao SW, Yip YL, Tsang CM, et al. Etiological factors of carcinoma. Cancer 2019; 125: 79–89.
nasopharyngeal carcinoma. Oral Oncol 2014; 50: 330–8. 46 Liu LT, Tang LQ, Chen QY, et al. The prognostic value of plasma
26 Liu Z, Chang ET, Liu Q, et al. Oral hygiene and risk of epstein-barr viral dna and tumor response to neoadjuvant
nasopharyngeal carcinoma—a population-based case-control study chemotherapy in advanced-stage nasopharyngeal carcinoma.
in China. Cancer Epidemiol Biomarkers Prev 2016; 25: 1201–7. Int J Radiat Oncol Biol Phys 2015; 93: 862–69.
27 Liu Z, Chang ET, Liu Q, et al. Quantification of familial risk of 47 Leung SF, Chan KC, Ma BB, et al. Plasma Epstein-Barr viral DNA
nasopharyngeal carcinoma in a high-incidence area. Cancer 2017; load at midpoint of radiotherapy course predicts outcome in
123: 2716–25. advanced-stage nasopharyngeal carcinoma. Ann Oncol 2014;
28 Chang ET, Liu Z, Hildesheim A, et al. active and passive smoking 25: 1204–08.
and risk of nasopharyngeal carcinoma: a population-based 48 Chan ATC, Hui EP, Ngan RKC, et al. Analysis of plasma
case-control study in southern China. Am J Epidemiol 2017; Epstein-Barr virus DNA in nasopharyngeal cancer after
185: 1272–80. chemoradiation to identify high-risk patients for adjuvant
29 Coghill AE, Hsu WL, Pfeiffer RM, et al. Epstein-Barr virus serology chemotherapy: a randomized controlled trial. J Clin Oncol 2018;
as a potential screening marker for nasopharyngeal carcinoma JCO2018777847.
among high-risk individuals from multiplex families in Taiwan. 49 Le QT, Zhang Q, Cao H, et al. An international collaboration to
Cancer Epidemiol BiomarkersPrev 2014; 23: 1213–19. harmonize the quantitative plasma Epstein-Barr virus DNA assay
30 Chan KCA, Woo JKS, King A, et al. Analysis of plasma Epstein-Barr for future biomarker-guided trials in nasopharyngeal carcinoma.
virus DNA to Screen for Nasopharyngeal Cancer. New Engl J Med Clin Cancer Res 2013; 19: 2208–15.
2017; 377: 513–22. 50 Kim KY, Le QT, Yom SS, et al. Current state of pcr-based
31 Amin MB, American Joint Committee on Cancer. AJCC cancer epstein-barr virus dna testing for nasopharyngeal cancer.
staging manual. 8th ed. New York: Springer; 2017. J Natl Cancer Inst 2017; 109(4).
32 Tang LL, Chen YP, Mao YP, et al. Validation of the 8th edition of 51 Liao XB, Mao YP, Liu LZ, et al. How does magnetic resonance
the uicc/ajcc staging system for nasopharyngeal carcinoma from imaging influence staging according to AJCC staging system for
endemic areas in the intensity-modulated radiotherapy era. nasopharyngeal carcinoma compared with computed tomography?
J Natl Compr Canc Netw 2017; 15: 913–9. Int J Radiat Oncol Biol Physics 2008; 72: 1368–77.
33 Tang LQ, Li CF, Li J, et al. Establishment and validation of 52 Chen WS, Li JJ, Hong L, Xing ZB, Wang F, Li CQ. Comparison of
prognostic nomograms for endemic nasopharyngeal carcinoma. MRI, CT and 18F-FDG PET/CT in the diagnosis of local and
J Natl Cancer Inst 2016; 108(1). metastatic of nasopharyngeal carcinomas: an updated meta analysis
of clinical studies. Am J Transl Res 2016; 8: 4532–47.
34 Jiang W, Liu N, Chen XZ, et al. Genome-wide identification of a
methylation gene panel as a prognostic biomarker in 53 Chua ML, Ong SC, Wee JT, et al. Comparison of 4 modalities for
nasopharyngeal carcinoma. Mol Cancer Ther 2015; 14: 2864–73. distant metastasis staging in endemic nasopharyngeal carcinoma.
Head Neck 2009; 31: 346–54.
35 Liu N, Chen NY, Cui RX, et al. Prognostic value of a microRNA
signature in nasopharyngeal carcinoma: a microRNA expression 54 Peng H, Chen L, Tang LL, et al. Significant value of (18)F-FDG-PET/
analysis. Lancet Oncol 2012; 13: 633–41. CT in diagnosing small cervical lymph node metastases in patients
with nasopharyngeal carcinoma treated with intensity-modulated
36 Tang XR, Li YQ, Liang SB, et al. Development and validation of
radiotherapy. Chin J Cancer 2017; 36: 95.
a gene expression-based signature to predict distant metastasis
in locoregionally advanced nasopharyngeal carcinoma: 55 Wei J, Pei S, Zhu X. Comparison of 18F-FDG PET/CT, MRI and
a retrospective, multicentre, cohort study. Lancet Oncol 2018; SPECT in the diagnosis of local residual/recurrent nasopharyngeal
19: 382–93. carcinoma: A meta-analysis. Oral Oncol 2016; 52: 11–7.
37 Fang W, Zhang J, Hong S, et al. EBV-driven LMP1 and 56 Chan SC, Yeh CH, Yen TC, et al. Clinical utility of simultaneous
IFN-gamma up-regulate PD-L1 in nasopharyngeal carcinoma: whole-body (18)F-FDG PET/MRI as a single-step imaging modality
implications for oncotargeted therapy. Oncotarget 2014; in the staging of primary nasopharyngeal carcinoma.
5: 12189–202. Eur J Nucl Med Mol Imaging 2018; 45: 1297–308.
38 Lee VH, Lo AW, Leung CY, et al. Correlation of PD-L1 expression of 57 Lin P, Min M, Lee M, et al. Prognostic utility of (18)F-FDG PET-CT
tumor cells with survival outcomes after radical intensity-modulated performed prior to and during primary radiotherapy for
radiation therapy for non-metastatic nasopharyngeal carcinoma. nasopharyngeal carcinoma: Index node is a useful prognostic
PLoS One 2016; 11: e0157969. imaging biomarker site. Radiother Oncol 2016; 120: 87–91.
39 Zhu Q, Cai MY, Chen CL, et al. Tumor cells PD-L1 expression as a 58 Chen YH, Chang KP, Chu SC, et al. Value of early evaluation of
favorable prognosis factor in nasopharyngeal carcinoma patients treatment response using (18)F-FDG PET/CT parameters and the
with pre-existing intratumor-infiltrating lymphocytes. Epstein-Barr virus DNA load for prediction of outcome in patients
Oncoimmunology 2017; 6: e1312240. with primary nasopharyngeal carcinoma.
Eur J Nucl Med Mol Imaging 2019; 46: 650–60.
40 Larbcharoensub N, Mahaprom K, Jiarpinitnun C, et al.
Characterization of PD-L1 and PD-1 expression and CD8+ 59 Gillies RJ, Kinahan PE, Hricak H. Radiomics: images are more than
tumor-infiltrating lymphocyte in Epstein-Barr virus-associated pictures, they are data. Radiology 2016; 278: 563–77.
nasopharyngeal carcinoma. Am J Clin Oncol 2018; 41: 1204–10. 60 Zhang B, Tian J, Dong D, et al. Radiomics features of
41 Ono T, Azuma K, Kawahara A, et al. Prognostic stratification of multiparametric mri as novel prognostic factors in advanced
patients with nasopharyngeal carcinoma based on tumor immune nasopharyngeal carcinoma. Clin Cancer Res 2017; 23: 4259–69.

microenvironment. Head Neck 2018; 40: 2007–19. 61 Pow EH, Kwong DL, McMillan AS, et al. Xerostomia and quality of
42 Wang YQ, Chen YP, Zhang Y, et al. Prognostic significance of life after intensity-modulated radiotherapy vs. conventional
tumor-infiltrating lymphocytes in nondisseminated radiotherapy for early-stage nasopharyngeal carcinoma: initial report
nasopharyngeal carcinoma: a large-scale cohort study. Int J Cancer on a randomized controlled clinical trial. Int J Radiat Oncol Biol Phys
2018; 142: 2558–66. 2006; 66: 981–91.
43 Xu C, Chen YP, Liu X, et al. Establishing and applying nomograms 62 Kam MK, Leung SF, Zee B, et al. Prospective randomized study of
based on the 8th edition of the UICC/AJCC staging system to select intensity-modulated radiotherapy on salivary gland function in
patients with nasopharyngeal carcinoma who benefit from early-stage nasopharyngeal carcinoma patients. J Clin Oncol 2007;
induction chemotherapy plus concurrent chemoradiotherapy. 25: 4873–9.
Oral Oncol 2017; 69: 99–107. 63 Peng G, Wang T, Yang KY, et al. A prospective, randomized study
44 Lee VH, Kwong DL, Leung TW, et al. The addition of pretreatment comparing outcomes and toxicities of intensity-modulated
plasma Epstein-Barr virus DNA into the eighth edition of radiotherapy vs. conventional two-dimensional radiotherapy for the
nasopharyngeal cancer TNM stage classification. Int J Cancer 2019; treatment of nasopharyngeal carcinoma. Radiother Oncol 2012;
144: 1713–22. 104: 286–93.

www.thelancet.com Vol 394 July 6, 2019 77


Seminar

64 Zhang B, Mo Z, Du W, Wang Y, Liu L, Wei Y. Intensity-modulated 83 Chen Y, Sun Y, Liang SB, et al. Progress report of a randomized trial
radiation therapy versus 2D-RT or 3D-CRT for the treatment of comparing long-term survival and late toxicity of concurrent
nasopharyngeal carcinoma: a systematic review and meta-analysis. chemoradiotherapy with adjuvant chemotherapy versus
Oral Oncol 2015; 51: 1041–46. radiotherapy alone in patients with stage III to IVB nasopharyngeal
65 Co J, Mejia MB, Dizon JM. Evidence on effectiveness of carcinoma from endemic regions of China. Cancer 2013;
intensity-modulated radiotherapy versus 2-dimensional 119: 2230–38.
radiotherapy in the treatment of nasopharyngeal carcinoma: 84 Blanchard P, Lee A, Marguet S, et al. Chemotherapy and
meta-analysis and a systematic review of the literature. Head Neck radiotherapy in nasopharyngeal carcinoma: an update of the
2016; 38 (suppl 1): E2130–42. MAC-NPC meta-analysis. Lancet Oncol 2015; 16: 645–55.
66 Mao YP, Tang LL, Chen L, et al. Prognostic factors and failure 85 Chen L, Hu CS, Chen XZ, et al. Concurrent chemoradiotherapy
patterns in non-metastatic nasopharyngeal carcinoma after plus adjuvant chemotherapy versus concurrent chemoradiotherapy
intensity-modulated radiotherapy. Chin J Cancer 2016; 35: 103. alone in patients with locoregionally advanced nasopharyngeal
67 Lewis GD, Holliday EB, Kocak-Uzel E, et al. Intensity-modulated carcinoma: a phase 3 multicentre randomised controlled trial.
proton therapy for nasopharyngeal carcinoma: decreased radiation Lancet Oncol 2012; 13: 163–71.
dose to normal structures and encouraging clinical outcomes. 86 Chen L, Hu CS, Chen XZ, et al. Adjuvant chemotherapy in patients
Head Neck 2016; 38 (suppl 1): E1886–95. with locoregionally advanced nasopharyngeal carcinoma: Long-term
68 Kong L, Gao J, Hu J, et al. Phase I/II trial evaluating concurrent results of a phase 3 multicentre randomised controlled trial.
carbon-ion radiotherapy plus chemotherapy for salvage treatment of Eur J Cancer 2017; 75: 150–8.
locally recurrent nasopharyngeal carcinoma. Chin J Cancer 2016; 87 Peng H, Chen L, Zhang Y, et al. Prognostic value of the cumulative
35: 101. cisplatin dose during concurrent chemoradiotherapy in
69 Hu J, Bao C, Gao J, et al. Salvage treatment using carbon ion locoregionally advanced nasopharyngeal carcinoma: a secondary
radiation in patients with locoregionally recurrent nasopharyngeal analysis of a prospective phase III clinical trial. Oncologist 2016;
carcinoma: Initial results. Cancer 2018; 124: 2427–37. 21: 1369–76.
70 Lee AW, Ng WT, Pan JJ, et al. International guideline for the 88 Loong HH, Ma BB, Leung SF, et al. Prognostic significance of
delineation of the clinical target volumes (CTV) for nasopharyngeal the total dose of cisplatin administered during concurrent
carcinoma. Radiother Oncol 2018; 126: 25–36. chemoradiotherapy in patients with locoregionally advanced
71 Sun Y, Yu XL, Luo W, et al. Recommendation for a contouring nasopharyngeal carcinoma. Radiother Oncol 2012; 104: 300–04.
method and atlas of organs at risk in nasopharyngeal carcinoma 89 Lee AW, Tung SY, Ngan RK, et al. Factors contributing to the efficacy
patients receiving intensity-modulated radiotherapy. Radiother Oncol of concurrent-adjuvant chemotherapy for locoregionally advanced
2014; 110: 390–97. nasopharyngeal carcinoma: combined analyses of NPC-9901 and
72 Tao CJ, Yi JL, Chen NY, et al. Multi-subject atlas-based NPC-9902 Trials. Eur J Cancer 2011; 47: 656–66.
auto-segmentation reduces interobserver variation and improves 90 Lv JW, Qi ZY, Zhou GQ, et al. Optimal cumulative cisplatin dose in
dosimetric parameter consistency for organs at risk in nasopharyngeal carcinoma patients receiving additional induction
nasopharyngeal carcinoma: a multi-institution clinical study. chemotherapy. Cancer Sci 2018; 109: 751–63.
Radiother Oncol 2015; 115: 407–11. 91 Tang LQ, Chen DP, Guo L, et al. Concurrent chemoradiotherapy
73 Lin L, Dou Q, Jin YM, et al. Deep learning for automated with nedaplatin versus cisplatin in stage II-IVB nasopharyngeal
contouring of primary tumor volumes by mri for nasopharyngeal carcinoma: an open-label, non-inferiority, randomised phase 3 trial.
carcinoma. Radiology 2019: 291: 677–86. Lancet Oncol 2018; 19: 461–73.
74 Colevas AD, Yom SS, Pfister DG, et al. NCCN guidelines insights: 92 Chen QY, Wen YF, Guo L, et al. Concurrent chemoradiotherapy vs
head and neck cancers, version 1.2018. J Natl Compr Canc Netw radiotherapy alone in stage II nasopharyngeal carcinoma: phase III
2018; 16: 479–90. randomized trial. J Natl Cancer Inst 2011; 103: 1761–70.
75 Lin JC, Jan JS, Hsu CY, Liang WM, Jiang RS, Wang WY. Phase III 93 Xu C, Zhang LH, Chen YP, et al. Chemoradiotherapy versus
study of concurrent chemoradiotherapy versus radiotherapy alone radiotherapy alone in stage II nasopharyngeal carcinoma:
for advanced nasopharyngeal carcinoma: positive effect on overall a systemic review and meta-analysis of 2138 patients. J Cancer 2017;
and progression-free survival. J Clin Oncol 2003; 21: 631–37. 8: 287–97.
76 Chan AT, Leung SF, Ngan RK, et al. Overall survival after 94 Zhang L, Huang Y, Hong S, et al. Gemcitabine plus cisplatin versus
concurrent cisplatin-radiotherapy compared with radiotherapy fluorouracil plus cisplatin in recurrent or metastatic nasopharyngeal
alone in locoregionally advanced nasopharyngeal carcinoma. carcinoma: a multicentre, randomised, open-label, phase 3 trial.
J Natl Cancer Inst 2005; 97: 536–39. Lancet 2016; 388: 1883–92.
77 Kwong DL, Sham JS, Au GK, et al. Concurrent and adjuvant 95 Bocci G, Kerbel RS. Pharmacokinetics of metronomic
chemotherapy for nasopharyngeal carcinoma: a factorial study. chemotherapy: a neglected but crucial aspect. Nat Rev Clin Oncol
J Clin Oncol 2004; 22: 2643–53. 2016; 13: 659–73.
78 Wu X, Huang PY, Peng PJ, et al. Long-term follow-up of a phase III 96 Chen YL, Chang MC, Cheng WF. Metronomic chemotherapy and
study comparing radiotherapy with or without weekly oxaliplatin for immunotherapy in cancer treatment. Cancer Lett 2017; 400: 282–92.
locoregionally advanced nasopharyngeal carcinoma. Ann Oncol 97 Twu CW, Wang WY, Chen CC, et al. Metronomic adjuvant
2013; 24: 2131–36. chemotherapy improves treatment outcome in nasopharyngeal
79 Al-Sarraf M, LeBlanc M, Giri PG, et al. Chemoradiotherapy versus carcinoma patients with postradiation persistently detectable
radiotherapy in patients with advanced nasopharyngeal cancer: plasma Epstein-Barr virus deoxyribonucleic acid.
phase III randomized Intergroup study 0099. J Clin Oncol 1998; Int J Radiat Oncology Biol Phys 2014; 89: 21–29.
16: 1310–17. 98 Liu YC, Wang WY, Twu CW, et al. Prognostic impact of adjuvant
80 Wee J, Tan EH, Tai BC, et al. Randomized trial of radiotherapy chemotherapy in high-risk nasopharyngeal carcinoma patients.
versus concurrent chemoradiotherapy followed by adjuvant Oral Oncol 2017; 64: 15–21.
chemotherapy in patients with American Joint Committee on 99 Chua DT, Ma J, Sham JS, et al. Long-term survival after
Cancer/International Union against cancer stage III and IV cisplatin-based induction chemotherapy and radiotherapy for
nasopharyngeal cancer of the endemic variety. J Clin Oncol 2005; nasopharyngeal carcinoma: a pooled data analysis of two phase III
23: 6730–38. trials. J Clin Oncol 2005; 23: 1118–24.
81 Lee AW, Tung SY, Chua DT, et al. Randomized trial of radiotherapy 100 Ribassin-Majed L, Marguet S, Lee AW, et al. What is the best
plus concurrent-adjuvant chemotherapy vs radiotherapy alone for treatment of locally advanced nasopharyngeal carcinoma?
regionally advanced nasopharyngeal carcinoma. J Natl Cancer Inst An individual patient data network meta-analysis. J Clin Oncol 2016;
2010; 102: 1188–98. 35: 498–505.
82 Lee AW, Tung SY, Chan AT, et al. A randomized trial on addition of 101 Hui EP, Ma BB, Leung SF, et al. Randomized phase II trial of
concurrent-adjuvant chemotherapy and/or accelerated fractionation concurrent cisplatin-radiotherapy with or without neoadjuvant
for locally-advanced nasopharyngeal carcinoma. Radiother Oncol docetaxel and cisplatin in advanced nasopharyngeal carcinoma.
2011; 98: 15–22. J Clin Oncol 2009; 27: 242–49.

78 www.thelancet.com Vol 394 July 6, 2019


Seminar

102 Fountzilas G, Ciuleanu E, Bobos M, et al. Induction chemotherapy 119 Li YQ, Tian YM, Tan SH, et al. Prognostic Model for Stratification
followed by concomitant radiotherapy and weekly cisplatin versus of Radioresistant Nasopharynx Carcinoma to Curative Salvage
the same concomitant chemoradiotherapy in patients with Radiotherapy. J Clin Oncol 2018; 36: 891–99.
nasopharyngeal carcinoma: a randomized phase II study conducted 120 Leong YH, Soon YY, Lee KM, Wong LC, Tham IWK, Ho FCH.
by the Hellenic Cooperative Oncology Group (HeCOG) with Long-term outcomes after reirradiation in nasopharyngeal
biomarker evaluation. Ann Oncol 2012; 23: 427–35. carcinoma with intensity-modulated radiotherapy: A meta-analysis.
103 Tan T, Lim WT, Fong KW, et al. Concurrent chemo-radiation with or Head Neck 2018; 40: 622–31.
without induction gemcitabine, carboplatin, and paclitaxel: 121 Tian YM, Tian YH, Zeng L, et al. Prognostic model for survival of
a randomized, phase 2/3 trial in locally advanced nasopharyngeal local recurrent nasopharyngeal carcinoma with intensity-modulated
carcinoma. Int J Radiat Oncol Biol Phys 2015; 91: 952–60. radiotherapy. Br J Cancer 2014; 110: 297–303.
104 Huang PY, Zeng Q, Cao KJ, et al. Ten-year outcomes of a 122 Tsang RK, Wei WI. Salvage surgery for nasopharyngeal cancer.
randomised trial for locoregionally advanced nasopharyngeal World J Otorhinolaryngol Head Neck Surg 2015; 1: 34–43.
carcinoma: a single-institution experience from an endemic area. 123 Chan JY, Tsang RK, Wei WI. Morbidities after maxillary swing
Eur J Cancer 2015; 51: 1760–70. nasopharyngectomy for recurrent nasopharyngeal carcinoma.
105 Lee AW, Ngan RK, Tung SY, et al. Preliminary results of trial NPC-0501 Head Neck 2015; 37: 487–92.
evaluating the therapeutic gain by changing from concurrent-adjuvant 124 Chen MY, Wang SL, Zhu YL, et al. Use of a posterior pedicle nasal
to induction-concurrent chemoradiotherapy, changing from septum and floor mucoperiosteum flap to resurface the
fluorouracil to capecitabine, and changing from conventional to nasopharynx after endoscopic nasopharyngectomy for recurrent
accelerated radiotherapy fractionation in patients with locoregionally nasopharyngeal carcinoma. Head Neck 2012; 34: 1383–88.
advanced nasopharyngeal carcinoma. Cancer 2015; 121: 1328–38.
125 Zou X, Han F, Ma WJ, et al. Salvage endoscopic
106 Sun Y, Li WF, Chen NY, et al. Induction chemotherapy plus nasopharyngectomy and intensity-modulated radiotherapy versus
concurrent chemoradiotherapy versus concurrent conventional radiotherapy in treating locally recurrent
chemoradiotherapy alone in locoregionally advanced nasopharyngeal carcinoma. Head Neck 2015; 37: 1108–15.
nasopharyngeal carcinoma: a phase 3, multicentre, randomised
126 You R, Zou X, Wang SL, et al. New surgical staging system for
controlled trial. Lancet Oncol 2016; 17: 1509–20.
patients with recurrent nasopharyngeal carcinoma based on the
107 Li WF, Chen NY, Zhang N, et al. Concurrent chemoradiotherapy AJCC/UICC rTNM classification system. Eur J Cancer 2015;
with/without induction chemotherapy in locoregionally advanced 51: 1771–79.
nasopharyngeal carcinoma: Long-term results of phase 3
127 Chan OS, Ngan RK. Individualized treatment in stage IVC
randomized controlled trial. Int J Cancer 2019; 45: 295–305.
nasopharyngeal carcinoma. Oral Oncol 2014; 50: 791–97.
108 Cao SM, Yang Q, Guo L, et al. Neoadjuvant chemotherapy followed
128 Lee AW, Ng WT, Chan YH, Sze H, Chan C, Lam TH. The battle
by concurrent chemoradiotherapy versus concurrent
against nasopharyngeal cancer. Radiother Oncol 2012; 104: 272–78.
chemoradiotherapy alone in locoregionally advanced
nasopharyngeal carcinoma: a phase III multicentre randomised 129 Shen LJ, Wang SY, Xie GF, et al. Subdivision of M category for
controlled trial. Eur J Cancer 2017; 75: 14–23. nasopharyngeal carcinoma with synchronous metastasis: time to
expand the M categorization system. Chin J Cancer 2015; 34: 450–58.
109 Frikha M, Auperin A, Tao Y, et al. A randomized trial of induction
docetaxel-cisplatin-5FU followed by concomitant cisplatin-RT 130 Zou X, You R, Liu H, et al. Establishment and validation of M1
versus concomitant cisplatin-RT in nasopharyngeal carcinoma stage subdivisions for de novo metastatic nasopharyngeal
(GORTEC 2006-02). Ann Oncol 2018; 29: 731–36. carcinoma to better predict prognosis and guide treatment.
Eur J Cancer 2017; 77: 117–26.
110 Hong RL, Hsiao CF, Ting LL, et al. Final results of a randomized
phase III trial of induction chemotherapy followed by concurrent 131 Sun XS, Liu LT, Liu SL, et al. Identifying optimal candidates for
chemoradiotherapy versus concurrent chemoradiotherapy alone in local treatment of the primary tumor among patients with de novo
patients with stage IVA and IVB nasopharyngeal carcinoma-Taiwan metastatic nasopharyngeal carcinoma: a retrospective cohort study
Cooperative Oncology Group (TCOG) 1303 Study. Ann Oncol 2018; based on Epstein-Barr virus DNA level and tumor response to
29: 1972–79. palliative chemotherapy. BMC cancer 2019; 19: 92.
111 Chen YP, Tang LL, Yang Q, et al. Induction chemotherapy plus 132 Jin Y, Ye X, Shao L, et al. Serum lactic dehydrogenase strongly
concurrent chemoradiotherapy in endemic nasopharyngeal predicts survival in metastatic nasopharyngeal carcinoma treated
carcinoma: individual patient data pooled analysis of four with palliative chemotherapy. Eur J Cancer 2013; 49: 1619–26.
randomized trials. Clin Cancer Res 2018; 24: 1824–33. 133 Jin Y, Cai XY, Cai YC, et al. To build a prognostic score model
112 Kwong DL, Nicholls J, Wei WI, et al. The time course of histologic containing indispensible tumour markers for metastatic
remission after treatment of patients with nasopharyngeal nasopharyngeal carcinoma in an epidemic area. Eur J Cancer 2012;
carcinoma. Cancer 1999; 85: 1446–53. 48: 882–88.
113 Lin GW, Wang LX, Ji M, Qian HZ. The use of MR imaging to detect 134 Sun P, Chen C, Xia Y, et al. The Ratio of C-Reactive Protein/
residual versus recurrent nasopharyngeal carcinoma following albumin is a novel inflammatory predictor of overall survival in
treatment with radiation therapy. Eur J Radiol 2013; 82: 2240–46. cisplatin-based treated patients with metastatic nasopharyngeal
carcinoma. Dis Markers 2017; 2017: 6570808.
114 Comoretto M, Balestreri L, Borsatti E, Cimitan M, Franchin G,
Lise M. Detection and restaging of residual and/or recurrent 135 Chee J, Loh KS, Tham I, et al. Prognostic stratification of patients
nasopharyngeal carcinoma after chemotherapy and radiation with metastatic nasopharyngeal carcinoma using a clinical and
therapy: comparison of MR imaging and FDG PET/CT. Radiology biochemical scoring system. J Cancer Res Clin Oncol 2017;
2008; 249: 203–11. 143: 2563–70.
115 Wang WY, Twu CW, Chen HH, et al. Long-term survival analysis of 136 Shen L, Li W, Wang S, et al. Image-based multilevel subdivision of
nasopharyngeal carcinoma by plasma Epstein-Barr virus DNA M1 category in TNM staging system for metastatic nasopharyngeal
levels. Cancer 2013; 119: 963–70. carcinoma. Radiology 2016; 280: 805–14.
116 Zhang Y, Tang LL, Li YQ, Liu X, Liu Q, Ma J. Spontaneous 137 Jin Y, Shi YX, Cai XY, et al. Comparison of five cisplatin-based
remission of residual post-therapy plasma Epstein-Barr virus DNA regimens frequently used as the first-line protocols in metastatic
and its prognostic implication in nasopharyngeal carcinoma: nasopharyngeal carcinoma. J Cancer Res Clin Oncol 2012;
a large-scale, big-data intelligence platform-based analysis. 138: 1717–25.
Int J Cancer 2019; 144: 2313–19. 138 Wang TL, Tan YO. Cisplatin and 5-fluorouracil continuous infusion
117 Zhang MX, Li J, Shen GP, et al. Intensity-modulated radiotherapy for metastatic nasopharyngeal carcinoma. Ann Acad Med Singapore
prolongs the survival of patients with nasopharyngeal carcinoma 1991; 20: 601–03.
compared with conventional two-dimensional radiotherapy: 139 Choo R, Tannock I. Chemotherapy for recurrent or metastatic
a 10-year experience with a large cohort and long follow-up. carcinoma of the nasopharynx. A review of the Princess Margaret
Eur J Cancer 2015; 51: 2587–95. Hospital experience. Cancer 1991; 68: 2120–24.
118 Liu YP, Li H, You R, et al. Surgery for isolated regional failure in 140 Lee AW, Ma BB, Ng WT, Chan AT. Management of nasopharyngeal
nasopharyngeal carcinoma after radiation: Selective or carcinoma: current practice and future perspective. J Clin Oncol
comprehensive neck dissection. Laryngoscope 2019; 129: 387–95. 2015; 33: 3356–64.

www.thelancet.com Vol 394 July 6, 2019 79


Seminar

141 Zhang L, Huang Y, Hong S, et al. Gemcitabine plus cisplatin versus 147 Iglesia MD, Parker JS, Hoadley KA, Serody JS, Perou CM,
fluorouracil plus cisplatin in recurrent or metastatic nasopharyngeal Vincent BG. Genomic analysis of immune cell infiltrates across
carcinoma: a multicentre, randomised, open-label, phase 3 trial. 11 tumor types. J Natl Cancer Inst 2016; 108.
Lancet 2016; 388: 1883–92. 148 Roh W, Chen PL, Reuben A, et al. Integrated molecular analysis of
142 Hsu C, Lee SH, Ejadi S, et al. Safety and antitumor activity of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals
pembrolizumab in patients with programmed death-ligand markers of response and resistance. Sci Transl Med 2017; 9.
1-positive nasopharyngeal carcinoma: results of the KEYNOTE-028 149 Davoli T, Uno H, Wooten EC, Elledge SJ. Tumor aneuploidy
study. J Clin Oncol 2017; 35: 4050–56. correlates with markers of immune evasion and with reduced
143 Ma BBY, Lim WT, Goh BC, et al. Antitumor activity of nivolumab in response to immunotherapy. Science 2017; 355.
recurrent and metastatic nasopharyngeal carcinoma: an international, 150 Thorsson V, Gibbs DL, Brown SD, et al. The immune landscape of
multicenter study of the Mayo Clinic Phase 2 Consortium (NCI-9742). cancer. Immunity 2018; 48: 812–30.e14.
J Clin Oncol 2018; 36: 1412–18. 151 Chen YP, Wang YQ, Lv JW, et al. Identification and validation of
144 Fang W, Yang Y, Ma Y, et al. Camrelizumab (SHR-1210) alone or in novel microenvironment-based immune molecular subgroups of
combination with gemcitabine plus cisplatin for nasopharyngeal head and neck squamous cell carcinoma: implications for
carcinoma: results from two single-arm, phase 1 trials. Lancet Oncol immunotherapy. Ann Oncol 2019; 30: 68–75.
2018; 19: 1338–50.
145 Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. © 2019 Elsevier Ltd. All rights reserved.
Mutational landscape determines sensitivity to PD-1 blockade in
non-small cell lung cancer. Science 2015; 348: 124–28.
146 Schumacher TN, Schreiber RD. Neoantigens in cancer
immunotherapy. Science 2015; 348: 69–74.

80 www.thelancet.com Vol 394 July 6, 2019

Potrebbero piacerti anche