You are on page 1of 12

Articles

Development and validation of a gene expression-based


signature to predict distant metastasis in locoregionally
advanced nasopharyngeal carcinoma: a retrospective,
multicentre, cohort study
Xin-Ran Tang*, Ying-Qin Li*, Shao-Bo Liang*, Wei Jiang*, Fang Liu*, Wen-Xiu Ge, Ling-Long Tang, Yan-Ping Mao, Qing-Mei He, Xiao-Jing Yang,
Yuan Zhang, Xin Wen, Jian Zhang, Ya-Qin Wang, Pan-Pan Zhang, Ying Sun, Jing-Ping Yun, Jing Zeng, Li Li, Li-Zhi Liu, Na Liu†, Jun Ma†

Summary
Background Gene expression patterns can be used as prognostic biomarkers in various types of cancers. We aimed to Lancet Oncol 2018
identify a gene expression pattern for individual distant metastatic risk assessment in patients with locoregionally Published Online
advanced nasopharyngeal carcinoma. February 7, 2018
http://dx.doi.org/10.1016/
S1470-2045(18)30080-9
Methods In this multicentre, retrospective, cohort analysis, we included 937 patients with locoregionally advanced
See Online/Comment
nasopharyngeal carcinoma from three Chinese hospitals: the Sun Yat-sen University Cancer Center (Guangzhou, China), http://dx.doi.org/10.1016/
the Affiliated Hospital of Guilin Medical University (Guilin, China), and the First People’s Hospital of Foshan (Foshan, S1470-2045(18)30085-8
China). Using microarray analysis, we profiled mRNA gene expression between 24 paired locoregionally advanced *Contributed equally to this
nasopharyngeal carcinoma tumours from patients at Sun Yat-sen University Cancer Center with or without distant Article
metastasis after radical treatment. Differentially expressed genes were examined using digital expression profiling in a †Senior authors contributed
training cohort (Guangzhou training cohort; n=410) to build a gene classifier using a penalised regression model. We equally to this work

validated the prognostic accuracy of this gene classifier in an internal validation cohort (Guangzhou internal validation Sun Yat-sen University Cancer
Center, State Key Laboratory of
cohort, n=204) and two external independent cohorts (Guilin cohort, n=165; Foshan cohort, n=158). The primary
Oncology in South China,
endpoint was distant metastasis-free survival. Secondary endpoints were disease-free survival and overall survival. Collaborative Innovation
Center of Cancer Medicine,
Findings We identified 137 differentially expressed genes between metastatic and non-metastatic locoregionally Guangzhou, China
(X-R Tang MD, Y-Q Li PhD,
advanced nasopharyngeal carcinoma tissues. A distant metastasis gene signature for locoregionally advanced
S-B Liang MD, L-L Tang PhD,
nasopharyngeal carcinoma (DMGN) that consisted of 13 genes was generated to classify patients into high-risk and Y-P Mao PhD, Q-M He MD,
low-risk groups in the training cohort. Patients with high-risk scores in the training cohort had shorter distant X-J Yang BS, Y Zhang MD,
metastasis-free survival (hazard ratio [HR] 4·93, 95% CI 2·99–8·16; p<0·0001), disease-free survival (HR 3·51, X Wen MD, J Zhang MD,
Y-Q Wang MD, P-P Zhang MD,
2·43–5·07; p<0·0001), and overall survival (HR 3·22, 2·18–4·76; p<0·0001) than patients with low-risk scores. The Prof Y Sun PhD, Prof J-P Yun PhD,
prognostic accuracy of DMGN was validated in the internal and external cohorts. Furthermore, among patients with J Zeng PhD, Prof L Li PhD,
low-risk scores in the combined training and internal cohorts, concurrent chemotherapy improved distant metastasis- L-Z Liu PhD, N Liu PhD,
free survival compared with those patients who did not receive concurrent chemotherapy (HR 0·40, 95% CI Prof J Ma MD); Department of
Radiation Oncology, The
0·19–0·83; p=0·011), whereas patients with high-risk scores did not benefit from concurrent chemotherapy (HR First People’s Hospital of
1·03, 0·71–1·50; p=0·876). This was also validated in the two external cohorts combined. We developed a nomogram Foshan, Foshan, China
based on the DMGN and other variables that predicted an individual’s risk of distant metastasis, which was (F Liu PhD, S-B Liang);
strengthened by adding Epstein–Barr virus DNA status. Department of Radiation
Oncology, Affiliated Hospital
of Guilin Medical University,
Interpretation The DMGN is a reliable prognostic tool for distant metastasis in patients with locoregionally advanced Guilin, China (W Jiang PhD); and
nasopharyngeal carcinoma and might be able to predict which patients benefit from concurrent chemotherapy. It has Department of Probability and
the potential to guide treatment decisions for patients at different risk of distant metastasis. Statistics, School of
Mathematical Sciences,
South China Normal University,
Funding The National Natural Science Foundation of China, the National Science & Technology Pillar Program Guangzhou, China
during the Twelfth Five-year Plan Period, the Natural Science Foundation of Guang Dong Province, the National Key (W-X Ge PhD)
Research and Development Program of China, the Innovation Team Development Plan of the Ministry of Education, Correspondence to:
the Health & Medical Collaborative Innovation Project of Guangzhou City, China, and the Program of Introducing Prof Jun Ma, Sun Yat-sen
University Cancer Center, State
Talents of Discipline to Universities. Key Laboratory of Oncology in
South China, Collaborative
Introduction standard treatment for patients with locoregionally Innovation Center of Cancer
Nasopharyngeal carcinoma has a unique geographical advanced nasopharyngeal carcinoma is radiotherapy Medicine, 651 Dongfeng Road
East, Guangzhou 510060, China
distribution, with the highest incidence in southeast Asia.1 combined with chemotherapy.3 With the use of intensity- majun2@mail.sysu.edu.cn
Of the 87 000 newly diagnosed cases annually, more than modulated radiotherapy and combined chemoradiotherapy,
70% of patients with nasopharyngeal carcinoma are locoregional control has improved substantially and distant
classified as having locoregionally advanced disease.2 The metastasis is now the main reason for treatment failure.4

www.thelancet.com/oncology Published online February 7, 2018 http://dx.doi.org/10.1016/S1470-2045(18)30080-9 1


Articles

Research in context
Evidence before this study predicts risk of distant metastasis and which patients are most
We searched PubMed up to Aug 23, 2017, for research articles likely to benefit from concurrent chemotherapy in locoregionally
containing the terms “gene expression profiling AND advanced nasopharyngeal carcinoma and that the DMGN is
locoregionally advanced nasopharyngeal carcinoma AND more accurate than clinicopathological risk factors alone. We
prognosis”, without date or language restrictions. Our search further developed a nomogram comprising DMGN, N stage, sex,
did not identify any previous high-throughput studies that had and serum lactate dehydrogenase and C-reactive protein
investigated the potential prognostic role of gene-expression concentrations that predicted distant metastasis-free survival in
profiles in locoregionally advanced nasopharyngeal carcinoma. the training cohort and was validated both internally and
externally.
Added value of this study
We did a multicentre, retrospective study to test the ability of Implications of all the available evidence
mRNA expression profiles of locoregionally advanced The DMGN is a reliable prognostic tool for distant metastasis
nasopharyngeal carcinoma tumours to predict the risk of distant in patients with locoregionally advanced nasopharyngeal
metastasis at the time of diagnosis. Our study included carcinoma and directly quantifies mRNA expression from
937 patients and is therefore, to our knowledge, the largest paraffin-embedded nasopharyngeal carcinoma tissues based
biomarker discovery project to be done in locoregionally on a commercially available mRNA NanoString platform,
advanced nasopharyngeal carcinoma. We developed and making it easy to implement in clinical practice. We believe
validated a distant metastasis gene signature for locoregionally that future prospective validation combining the DMGN with
advanced nasopharyngeal carcinoma (DMGN) that is based on Epstein–Barr virus DNA status will improve the basis for clinical
the expression of 13 genes and can be used as a molecular decision making for patients with locoregionally advanced
classifier for distant metastasis. We show that this signature nasopharyngeal carcinoma.

The tumour–node–metastasis (TNM) staging system is responses in various types of cancers.10–13 In nasopharyngeal
the key determinant for prognostic prediction and risk carcinoma, recent evidence also indicates that certain
stratification for treatment decisions. Even though gene-expression patterns might be used as molecular
patients with locoregionally advanced nasopharyngeal markers that allow early diagnosis and subgroup
carcinoma with the same TNM stage receive similar classification.14–16 However, how expression patterns might
treatments, their clinical outcomes vary greatly. differ between patients with different clinical outcomes,
Approximately 30–40% of patients with locoregionally especially distant metastasis, has not been established.
advanced nasopharyngeal carcinoma eventually develop We did a study of global gene expression with the aim of
distant metastasis after undergoing radical treatment.5 identifying and validating a gene-expression signature that
Hence, the current anatomical-based staging system is predicts distant metastasis in patients with locoregionally
not sufficient to predict which patients will develop distant advanced nasopharyngeal carcinoma. Moreover, we
metastasis. The differences in prognosis might be combined both genomic and clinical variables to generate
attributed to biological heterogeneity, and molecular a nomogram model with more accurate prediction than
investigation could provide biomarkers for predicting clinical risk factors for distant metastasis.
distant metastasis and guiding treatment decisions for
patients in different risk groups. Great efforts have been Methods
made to search for molecular biomarkers for locoregionally Clinical specimens and study design
advanced nasopharyngeal carcinoma, such as Epstein– We retrospectively collected 955 paraffin-embedded biopsy
Barr virus DNA (EBV DNA) concentration, lactate tissues from patients with stage III–IVa locoregionally
dehydrogenase (LDH) con­centration, and miRNAs.6–8 A advanced nasopharyngeal carcinoma, of which 937 passed
See Online for appendix randomised controlled trial using EBV DNA status to quality control for the final analysis (appendix p 22).
direct chemoradiotherapy in nasopharyngeal carcinoma is 614 samples were obtained between Jan 14, 2006, and
ongoing.9 However, new biomarkers that reflect tumour Dec 27, 2009, from patients treated at the Sun Yat-sen
heterogeneity still need to be identified to guide individual University Cancer Center (Guangzhou, China). Computer-
treatment for patients. generated random numbers were used to assign these
Comparisons of the global gene-expression profiles of samples into a training cohort (Guangzhou training
different cell types, developmental stages, and disease cohort) consisting of 410 samples and an internal validation
states could promote the discovery of characteristic gene cohort (Guangzhou internal validation cohort) consisting
signatures that might be correlated with functionally of 204 samples. 165 samples collected from the Affiliated
important states. Emerging evidence demonstrates that Hospital of Guilin Medical College (Guilin, China; Guilin
gene-expression profiling of formalin-fixed paraffin- external validation cohort) between Feb 26, 2007, and
embedded (FFPE) tissues is useful for molecular July 23, 2011, and 158 samples collected from the First
classification and prediction for prognoses and treatment People’s Hospital of Foshan (Foshan, China; Foshan

2 www.thelancet.com/oncology Published online February 7, 2018 http://dx.doi.org/10.1016/S1470-2045(18)30080-9


Articles

external validation cohort) between April 27, 2010, and subgroups of patients, then validate the findings using
March 3, 2014, were separately used as two independent low-throughput methods in larger populations.18,19 Based
external validation cohorts. All samples were reassessed by on this approach, we first compared gene expression
two pathologists (J-PY and JZ), and found to contain more profiles across 24 paired locoregionally advanced
than 70% tumour cells. None of the patients had received nasopharyngeal carcinoma tumour tissues from
any anti-tumour therapy before biopsy sampling. 48 patients at Sun Yat-sen University Cancer Center with
All patients underwent staging MRI, and patients with or without distant metastasis after curative treatment
contraindications for MRI received contrast-enhanced CT using the Affymetrix Human Gene 2.0 ST microarray
scans. Two radiologists (L-ZL and LL) separately (appendix pp 5, 6). Tissues were strictly matched by sex,
reassessed all MRI and CT scans, and any disagreements age, T stage, N stage, radiotherapy, and chemotherapy to
were resolved by consensus. We restaged all patients rule out the influence of these parameters on metastasis.
according to the 8th edition of the American Joint We then analysed the differentially expressed genes using
Committee on Cancer Staging Manual.17 Radiotherapy digital expression profiling (NanoString nCounter
dose and chemotherapy regimens are detailed in the system, NanoString Technologies, Seattle, WA, USA),
appendix (p 2). The institutional ethical review boards of and generated a distant metastasis gene signature for
all included hospitals approved this retrospective analysis locoregionally advanced nasopharyngeal carcinoma
of anonymous data, and the requirement for informed (DMGN) in the training cohort. Raw NanoString counts
consent was waived by the ethics review boards. for each gene within each experiment were subjected to
technical normalisation before biological normalisation
Procedures (appendix p 3). We used the counts obtained for the
Total RNA was extracted from FFPE samples using the appropriate positive control probe set, which consisted of
QIAGEN FFPE RNeasy kit (QIAGEN GmbH, Hilden, five housekeeping genes included in the NanoString
Germany). RNA was analysed using a Nanodrop 2000 CodeSet (appendix pp 7–10). The normalised data were
spectrophotometer (ThermoFisher Scientific, Waltham, then log2-transformed and used as the input for class
MA, USA), and RNA integrity numbers were determined prediction analysis. The methods used to select reference
to evaluate RNA integration using an Agilent genes and apply quality control criteria for the NanoString
Bioanalyzer 2100 (Agilent Technologies, Santa Clara, CA, data are described in detail in the appendix (pp 2, 3).
USA). Total RNA was amplified using an Ovation FFPE We used a penalised logistic model to select variables for
WTA System (NuGEN, San Carlos, CA, USA), and an constructing a prediction model. The R package glmnet
Encore Biotin Module (NuGEN) was used for was used to perform the least absolute shrinkage and
fragmentation and labelling. selection operator (LASSO) on the logistic regression
Total RNA was hybridised to Affymetrix Human model. Bootstrap methods were used to test the robustness
Gene 2.0 ST GeneChips (Affymetrix, Santa Clara, CA, of the significant candidate genes that were chosen by the
USA) using a GeneChip Hybridization, Wash and Stain penalised logistic model. Then, we calculated the area
Kit (Affymetrix) in a Hybridization Oven 645 (Affymetrix) under the receiver-operator characteristic (ROC) curve
and Fluidics Station 450 (Affymetrix) according to the (AUC) to determine how many candidate genes should be
manufacturer’s instructions. All slides were scanned using chosen (appendix p 23), and we constructed the prediction
a GeneChip Scanner 3000 (Affymetrix) and Command model with the coefficients weighted by the Cox model in
Console Software 3.1 (Affymetrix) with default settings. the training cohort.20 We used X-tile software (version 3.6.1;
For data processing, raw data were normalised using the Yale University, New Haven, CT, USA) in the training
oligo package in R with Robust Multi-array Analysis cohort to find the optimal cutoff value. The thresholds for
(RMA). We implemented Bayes statistics using empirical the scores that were outputted from the predictive model
Bayes (eBayes) in the limma package to compute which that was used to separate patients into low-risk and high-
probes were significantly differentially expressed between risk groups were defined as the score that produced the
metastatic and non-metastatic samples. Because RNA largest χ² value in the Mantel–Cox test.21
obtained from FFPE samples is highly and randomly Our primary endpoint was distant metastasis-free
fragmented, we used Fisher’s exact test to verify whether a survival. Secondary endpoints were disease-free survival
transcript was indeed significantly differentially expressed. and overall survival. We calculated distant metastasis-free
Differentially expressed transcripts were characterised survival as the time from day 1 of concurrent
according to the following criteria: an empirical fold- chemoradiotherapy or induction chemotherapy (if given)
change greater than 1·5 and an eBayes test p value less to the date of first distant relapse; disease-free survival to
than 0·05. The methods for data analysis are described in the first relapse at any site, death from any cause, or the
detail in the appendix (p 2). date of the last follow-up visit, whichever occurred first;
and overall survival to death from any cause.
Statistical analysis Distant metastasis-free survival, overall survival, and
Expression profiling studies often use high-throughput disease-free survival were calculated using the
assays to screen differentially expressed profiles in small Kaplan-Meier method and the log-rank test, and hazard

www.thelancet.com/oncology Published online February 7, 2018 http://dx.doi.org/10.1016/S1470-2045(18)30080-9 3


Articles

ratios (HRs) were calculated using a univariate Cox specificity of the model for predicting distant metastasis.
regression analysis. The DMGN as a predictor for Statistical analyses were done in R (version 3.2.1) and
concurrent chemotherapy efficacy was analysed in the SPSS (version 22.0). All statistical tests were two-sided,
combined training and internal cohorts and validated in and p values of less than 0·05 were deemed significant.
a combination cohort of the two external cohorts.
We also did a multivariate Cox regression analysis using Data sharing
backward selection to test the independent significance of The key raw data have been uploaded onto the Research
different factors; the p value threshold was 0·1 (p>0·1) for Data Deposit public platform (RDD), with the approval
For microarray data see https:// removing non-significant variables from the analysis, and RDD number of RDDB2017000204. The microarray data
www.ncbi.nlm.nih.gov/geo/ marginally significant variables (0·05<p<0·1) remained in have been deposited online under accession number
the final Cox model. Covariates included DMGN (high risk GSE103611.
vs low risk), sex, age (≥45 years vs <45 years), N stage
(N2–3 vs N0–1), T stage (T3–4 vs T1–2), PET–CT (no vs yes), Role of the funding source
viral capsid antigen immunoglobulin (VCA-IgA; titre The sponsors had no role in the study design, data
≥1:80 vs <1:80), early antigen immunoglobulin (EA-IgA; collection, data analysis, data interpretation, or writing of
titre ≥1:10 vs <1:10), serum LDH (≥245 vs <245 U/L), the report. The corresponding authors had full access to
C-reactive protein (≥8·2 vs <8·2 mg/L), haemoglobin the data and had final responsibility for the decision to
(<120 vs ≥120 g/L), body-mass index (BMI; ≥18·5 vs submit the manuscript for publication.
<18·5 kg/m²), and concurrent chemotherapy (no vs yes).
We formulated nomograms using the rms package in Results
R, and included serum LDH and C-reactive protein in We included in our analysis 937 pretreatment,
the nomogram as they are usually included in most non-­­meta­static nasopharyngeal carcinoma specimens
prognostic models of nasopharyngeal carcinoma.22,23 We that were obtained from three academic institutions in
used the coefficients of the multivariable Cox regression China (figure 1). The table shows the clinical
model to generate nomograms. Calibration curves were characteristics of the patients in the Guangzhou training
assessed graphically by plotting the observed rates cohort (n=410), Guangzhou internal validation cohort
against the nomogram predicted probabilities and a (n=204), and two external validation cohorts (Guilin,
concordance index (C-index) was calculated via a n=165; Foshan, n=158). All patients under­went radical
bootstrap method with 1000 resamples. We investigated radiotherapy, and platinum-based chemo­ therapy was
the prognostic or predictive accuracy of clinical features administered to 516 (84%) of 614 patients in the
and the multi-gene-based classifier using ROC analysis.24 Guangzhou cohort, 154 (93%) of 165 in the Guilin cohort,
We calculated the AUC to assess the sensitivity and and 147 (93%) of 158 in the Foshan cohort (appendix
p 11). Median follow-up was 78·2 months (IQR 49·0–89·4)
for patients in the Guangzhou cohort, 68·2 months
Affymetrix array for global gene expression
24 LA-NPC samples from patients without
(49·9–84·4) for those in the Guilin cohort, and
metastasis after radical treatment 49·9 months (38·9–64·4) for those in the Foshan cohort.
24 LA-NPC samples from patients with distant In the microarray analysis, 137 mRNAs were found to be
metastasis after radical treatment
differentially expressed between 24 patients with non­ -­
metastatic nasopharyngeal carcinoma and 24 patients
137 differentially expressed mRNAs with nasopharyngeal carcinoma who developed distant
metastasis after treatment. We found 13 of these differ­
entially expressed genes were associated with distant
Quantification of mRNAs with NanoString
nCounter system metastasis-free survival in the training cohort (appendix
p 23). A risk score was calculated for each patient using a
formula derived from the expression levels of these 13 genes
DMGN risk score devlopment in Guangzhou weighted by their regression coefficient (appendix p 12):
training cohort (n=410)

Risk score = (0·1846 × expression of YBX3) –


Validation of risk score in three cohorts (0·3007 × expression of CBR3) – (0·1383 ×
expression of CXCL10) – (0·3661 × expression of
CLASP1) + (0·2381 × expression of DCTN1) –
(0·4004 × expression of FNDC3B) + (0·60 ×
Guangzhou internal validation Guilin external validation Foshan external validation
cohort (n=204) cohort (n=165) cohort (n=158)
expression of WSB2) + (0·1093 × expression of
LRIG1) – (0·1162 × expression of GRM4) + (0·1327
× expression of ANXA1) + (0·1485 × expression of
Figure 1: Study flow
LA-NPC=locoregionally advanced nasopharyngeal carcinoma. DMGN=distant metastasis gene signature for WNK1) + (0·0714 × expression of HDLBP) +
locoregionally advanced nasopharyngeal carcinoma. (0·1774 × expression of POLR2M)

4 www.thelancet.com/oncology Published online February 7, 2018 http://dx.doi.org/10.1016/S1470-2045(18)30080-9


Articles

Guangzhou training cohort (n=410) Guangzhou internal validation cohort Guilin external validation cohort Foshan external validation cohort
(n=204) (n=165) (n=158)
Patients (n) Low risk High risk Patients (n) Low risk High risk Patients (n) Low risk High risk Patients (n) Low risk High risk
Age (years)
<45 181 104 (57%) 77 (43%) 89 44 (49%) 45 (51%) 64 39 (60%) 25 (40%) 63 34 (54%) 29 (46%)
≥45 229 103 (45%) 126 (55%) 115 61 (50%) 54 (50%) 101 57 (56%) 44 (44%) 95 53 (56%) 42 (44%)
Sex
Male 314 147 (47%) 167 (53%) 153 75 (49%) 78 (51%) 121 62 (51%) 59 (49%) 121 63 (52%) 58 (48%)
Female 96 60 (63%) 36 (38%) 51 30 (59%) 21 (41%) 44 34 (77%) 10 (23%) 37 24 (65%) 13 (35%)
WHO pathological type
Undifferentiated 398 201 (51%) 197 (49%) 198 101 (51%) 97 (49%) 164 96 (59%) 68 (41%) 158 87 (55%) 71 (45%)
non-keratinising
Differentiated 12 6 (50%) 6 (50%) 6 4 (67%) 2 (33%) 1 0 (0%) 1 (100%) 0 0 (0%) 0 (0%)
non-keratinising
T stage
T1 15 6 (40%) 9 (60%) 9 4 (44%) 5 (56%) 4 3 (75%) 1 (25%) 12 5 (42%) 7 (58%)
T2 60 28 (47%) 32 (53%) 28 16 (57%) 12 (43%) 31 18 (58%) 13 (42%) 11 6 (55%) 5 (45%)
T3 253 138 (55%) 115 (45%) 118 65 (55%) 53 (45%) 84 46 (55%) 38 (45%) 75 45 (60%) 30 (40%)
T4 82 35 (43%) 47 (57%) 49 20 (41%) 29 (59%) 46 29 (63%) 17 (37%) 60 31 (52%) 29 (48%)
N stage
N0 44 23 (52%) 21 (48%) 17 7 (41%) 10 (59%) 12 7 (58%) 5 (42%) 16 7 (44%) 9 (56%)
N1 192 105 (55%) 87 (45%) 80 40 (50%) 40 (50%) 54 35 (65%) 19 (35%) 71 41 (48%) 30 (42%)
N2 103 48 (47%) 55 (53%) 65 38 (58%) 27 (42%) 82 46 (56%) 36 (44%) 62 34 (55%) 28 (45%)
N3 71 31 (44%) 40 (56%) 42 20 (48%) 22 (52%) 17 8 (47%) 9 (53%) 9 5 (56%) 4 (44%)
TNM stage
III 264 145 (55%) 119 (45%) 120 68 (57%) 52 (43%) 103 61 (59%) 42 (41%) 91 52 (57%) 39 (43%)
IV 146 62 (42%) 84 (58%) 84 37 (44%) 47 (56%) 62 35 (56%) 27 (44%) 67 35 (52%) 32 (48%)
VCA-IgA
<1:80 45 24 (53%) 21 (47%) 22 12 (55%) 10 (45%) 119 66 (55%) 53 (45%) NA NA NA
≥1:80 365 183 (50%) 182 (50%) 182 93 (51%) 89 (49%) 46 30 (65%) 16 (35%) NA NA NA
EA-IgA
<1:10 76 41 (54%) 35 (46%) 43 27 (63%) 16 (37%) 119 66 (55%) 53 (45%) NA NA NA
≥1:10 334 166 (50%) 168 (50%) 161 78 (48%) 83 (52%) 46 30 (65%) 16 (35%) NA NA NA
LDH concentration (U/L)
<245 385 198 (51%) 187 (49%) 183 92 (50%) 91 (50%) NA NA NA 145 78 (54%) 67 (46%)
≥245 25 9 (36%) 16 (64%) 21 13 (62%) 8 (38%) NA NA NA 13 9 (69%) 4 (31%)
C-reactive protein concentration (mg/L)
<8·20 339 171 (50%) 168 (50%) 167 90 (54%) 77 (46%) NA NA NA 145 79 (54%) 66 (46%)
≥8·20 71 36 (51%) 35 (49%) 37 15 (41%) 22 (59%) NA NA NA 13 8 (62%) 5 (39%)
Haemoglobin concentration (g/L)
<120 111 58 (52%) 53 (48%) 47 19 (40%) 28 (60%) 70 49 (70%) 21 (30%) 20 9 (45%) 11 (55%)
≥120 299 149 (50%) 150 (50%) 157 86 (55%) 71 (45%) 95 47 (49%) 48 (51%) 138 78 (57%) 60 (43%)
Body-mass index (kg/m²)
<18·5 21 13 (62%) 8 (38%) 8 4 (50%) 4 (50%) 17 14 (82%) 3 (18%) 21 11 (52%) 10 (48%)
≥18·5 389 194 (50%) 195 (50%) 196 101 (52%) 95 (48%) 148 82 (55%) 66 (45%) 137 76 (55%) 61 (45%)
Concurrent chemotherapy
Yes 204 100 (49%) 104 (51%) 122 59 (48%) 63 (52%) 77 45 (58%) 32 (42%) 127 65 (51%) 62 (49%)
No 206 107 (52%) 99 (48%) 82 46 (56%) 36 (44%) 88 51 (58%) 37 (42%) 31 22 (71%) 9 (29%)

Data are n (%) unless otherwise stated. TNM=tumour–node–metastasis.VCA-IgA=viral capsid antigen immunoglobulin A. EA-IgA=early antigen immunoglobulin A. LDH=serum lactate dehydrogenase. NA=not
available.

Table: Clinical characteristics of patients in the training, internal, and external validation cohorts

We used X-tile plots to generate an optimal cutoff (appendix p 24). This assay assigned
value (5·01) to separate patients into low-risk and 207 (50·5%) of 410 patients to the low-risk group and
high-risk groups in the Guangzhou training cohort 203 (49·5%) to the high-risk group. 5-year distant

www.thelancet.com/oncology Published online February 7, 2018 http://dx.doi.org/10.1016/S1470-2045(18)30080-9 5


Articles

A Guangzhou training cohort B Guangzhou internal validation cohort


Distant metastasis-free survival (%) 100

75

50

25 Low risk
High risk
HR 4·93 (95% CI 2·99–8·16) HR 2·98 (95% CI 1·60–5·55)
p<0·0001 p=0·00032
0
0 12 24 36 48 60 72 84 96 108 120 0 12 24 36 48 60 72 84 96 108 120
Number at risk
(number
censored)
Low risk 207 (0) 199 (4) 191 (2) 177 (7) 171 (6) 160 (10) 124 (36)78 (45) 28 (50) 7 (21) 0 (7) 105 (0) 100 (2) 94 (2) 88 (4) 82 (3) 72 (9) 61 (10) 43 (18) 17 (26) 0 (17) 0 (0)
High risk 203 (0) 178 (3) 155 (3) 140 (0) 124 (6) 103 (16) 81 (20) 49 (31) 23 (24) 5 (18) 0 (5) 99 (0) 84 (0) 73 (0) 66 (4) 60 (4) 56 (3) 39 (16) 21 (18) 9 (11) 3 (6) 0 (3)

C Guilin external validation cohort D Foshan external validation cohort


100
Distant metastasis-free survival (%)

75

50

25

HR 2·92 (95% CI 1·55–5·49) HR 3·62 (95% CI 1·43–9·19)


p=0·00050 p=0·0037
0
0 12 24 36 48 60 72 84 96 108 0 12 24 36 48 60 72 84
Time since treatment (months) Time since treatment (months)
Number at risk
(number
censored)
Low risk 96 (0) 90 (0) 89 (0) 85 (0) 82 (0) 74 (7) 55 (19) 36 (19) 10 (26) 0 (10) 87 (0) 82 (3) 75 (5) 73 (1) 47 (26) 26 (20) 7 (19) 0 (7)
High risk 69 (0) 62 (0) 50 (0) 45 (0) 41 (1) 32 (9) 19 (13) 10 (9) 0 (10) 0 (0) 71 (0) 69 (0) 60 (3) 47 (7) 35 (9) 23 (12) 7 (16) 0 (7)

Figure 2: Kaplan-Meier curves of distant metastasis-free survival according to the DMGN


(A) Guangzhou training cohort (n=410), (B) Guangzhou internal validation cohort (n=204), (C) Guilin external validation cohort (n=165), and (D) Foshan external
validation cohort (n=158). We calculated p values using the unadjusted log-rank test and hazard ratios using a univariate Cox regression analysis. DMGN=distant
metastasis gene signature for locoregionally advanced nasopharyngeal carcinoma. HR=hazard ratio.

metastasis-free survival was 63·3% (95% CI 56·1–69·6) metastasis-free survival (HR 2·98, 95% CI 1·60–5·55;
for the high-risk group and 90·9% (85·9–94·2) in the p=0·00032; figure 2B), disease-free survival (HR 2·14,
low-risk group (HR 4·93, 95% CI 2·99–8·16; p<0·0001; 1·30–3·54; p=0·0023; appendix p 25), and overall survival
figure 2A; appendix pp 13, 14). Patients with high-risk (2·00, 1·22–3·27; p=0·0049; appendix p 26). The 5-year
scores also had shorter disease-free survival (HR 3·51, distant metastasis-free survival, disease-free survival, and
95% CI 2·43–5·07; p<0·0001; appendix pp 13, 25) and overall survival in each risk group and the number of
overall survival (HR 3·22, 2·18–4·76; p<0·0001; appendix patients who had an event for each risk group are listed in
pp 13, 26) than patients with low-risk scores. The 5-year the appendix (pp 13, 14).
distant metastasis-free survival, disease-free survival, and In the Guilin external validation cohort, the DMGN
overall survival in each risk group and the number of classified 96 (58%) of 165 patients into the low-risk
patients who had an event for each risk group are listed in group and 69 (42%) patients into the high-risk group.
the appendix (pp 13, 14). Patients with high-risk scores had shorter distant
In the Guangzhou internal validation cohort, the metastasis-free survival (HR 2·92, 95% CI 1·55–5·49;
DMGN categorised 105 (51%) of 204 patients into the low- p=0·00050; figure 2C), disease-free survival (HR 2·12,
risk group and 99 patients (49%) into the high-risk group, 1·29–3·48; p=0·0024; appendix p 25), and overall
which were significantly different in terms of distant survival (HR 2·33, 1·35–4·04; p=0·0018; appendix p 26)

6 www.thelancet.com/oncology Published online February 7, 2018 http://dx.doi.org/10.1016/S1470-2045(18)30080-9


Articles

Events (n)/ Events (n)/ HR (95% CI) p value


patients (N) patients (N)
in patients with in patients without
risk factor risk factor

Sex (male vs female)


Guangzhou training cohort 83/314 13/96 2·15 (1·20–3·86) 0·010
Guangzhou internal cohort 43/153 5/51 3·19 (1·26–8·07) 0·014
Guilin external cohort 36/121 6/44 2·52 (1·06–5·99) 0·036
Foshan external cohort 19/121 4/37 1·63 (0·55–4·81) 0·374
Overall 181/709 28/228 2·33 (1·56–3·47) <0·0001

Age (years) (≥45 vs <45)


Guangzhou training cohort 64/229 32/181 1·65 (1·08–2·53) 0·020
Guangzhou internal cohort 24/115 24/89 0·76 (0·43–1·34) 0·339
Guilin external cohort 27/101 15/64 1·18 (0·63–2·22) 0·608
Foshan external cohort 15/95 8/63 1·40 (0·59–3·31) 0·440
Overall 130/540 79/397 1·26 (0·95–1·66) 0·112

T stage (T3–4 vs T1–2)


Guangzhou training cohort 73/335 23/75 0·65 (0·41–1·05) 0·075
Guangzhou internal cohort 38/167 10/37 0·85 (0·43–1·71) 0·653
Guilin external cohort 35/130 7/35 1·38 (0·61–3·11) 0·437
Foshan external cohort 19/135 4/23 0·87 (0·30–2·56) 0·801
Overall 165/767 44/170 0·82 (0·59–1·14) 0·241

N stage (N2–3 vs N0–1)


Guangzhou training cohort 51/174 45/236 1·72 (1·15–2·57) 0·0079
Guangzhou internal cohort 35/107 13/97 2·80 (1·48–5·29) 0·0015
Guilin external cohort 32/99 10/66 2·38 (1·17–4·85) 0·017
Foshan external cohort 15/71 8/87 2·44 (1·04–5·76) 0·041
Overall 133/451 76/486 2·11 (1·59–2·79) <0·0001

DMGN (high risk vs low risk)


Guangzhou training cohort 77/203 19/207 4·93 (2·99–8·16) <0·0001
Guangzhou internal cohort 34/99 14/105 2·98 (1·60–5·55) 0·00061
Guilin external cohort 27/69 15/96 2·92 (1·55–5·49) 0·0090
Foshan external cohort 17/71 6/87 3·62 (1·43–9·19) 0·0067
Overall 155/442 54/495 3·73 (2·74–5·09) <0·0001

Haemoglobin (g/L) (<120 vs ≥120)


Guangzhou training cohort 76/299 20/111 0·72 (0·44–1·17) 0·186
Guangzhou internal cohort 31/157 17/47 2·24 (1·24–4·05) 0·0077
Guilin external cohort 24/95 18/70 0·99 (0·54–1·82) 0·968
Foshan external cohort 19/138 4/20 1·69 (0·57–4·97) 0·341
Overall 150/689 59/248 1·14 (0·84–1·54) 0·395

BMI (kg/m2) (≥18·5 vs <18·5)


Guangzhou training cohort 91/389 5/21 0·98 (0·40–2·41) 0·960
Guangzhou internal cohort 45/196 3/8 0·64 (0·20–2·07) 0·456
Guilin external cohort 38/148 4/17 1·09 (0·39–3·06) 0·867
Foshan external cohort 20/137 3/21 1·11 (0·33–3·72) 0·872
Overall 194/870 15/67 1·01 (0·60–1·70) 0·982

LDH (U/L) (≥245 vs <245)


Guangzhou training cohort 12/25 84/385 2·74 (1·50–5·02) 0·0011
Guangzhou internal cohort 10/21 38/183 2·85 (1·42–5·74) 0·0033
Foshan external cohort 5/13 18/145 3·65 (1·35–9·85) 0·011
Overall 27/59 140/713 2·92 (1·93–4·41) <0·0001

C-reactive protein (mg/L) (≥8·2 vs <8·2)


Guangzhou training cohort 23/71 73/339 1·67 (1·05–2·67) 0·032
Guangzhou internal cohort 13/37 35/167 1·93 (1·02–3·65) 0·043
Foshan external cohort 4/13 19/145 3·16 (1·07–9·32) 0·037 Figure 3: Univariate
Overall 40/121 127/651 1·90 (1·33–2·71) 0·00041 association of DMGN and
clinicopathological
VCA-IgA (≥1:80 vs <1:80) characteristics with distant
Guangzhou training cohort 84/365 12/45 0·89 (0·48–1·62) 0·696
metastasis-free survival
Guangzhou internal cohort 45/182 3/22 2·00 (0·62–6·44) 0·245
Guilin external cohort 8/46 34/119 0·56 (0·26–1·21) 0·142 DMGN=distant metastasis
Overall 137/593 49/186 0·90 (0·63–1·21) 0·422 gene signature for
locoregionally advanced
EA-IgA (≥1:10 vs <1:10) nasopharyngeal carcinoma.
Guangzhou training cohort 80/334 16/76 1·22 (0·72–2·01) 0·460 BMI=body-mass index.
Guangzhou internal cohort 41/161 7/43 1·72 (0·77–3·82) 0·187
LDH=serum lactate
Guilin external cohort 8/46 34/119 0·56 (0·26–1·21) 0·142
Overall 129/541 57/238 1·02 (0·75–1·40) 0·879 dehydrogenase. VCA-IgA=viral
capsid antigen
immunoglobulin A.
0·125 0·25 0·5 1 2 4 8 EA-IgA=early antigen
immunoglobulin A.
Favours control Favours risk
HR=hazard ratio.

www.thelancet.com/oncology Published online February 7, 2018 http://dx.doi.org/10.1016/S1470-2045(18)30080-9 7


Articles

than patients with low-risk scores. Similarly, in the Figure 3 shows univariate analysis of distant metastasis-
Foshan external validation cohort, 87 (55%) of 158 patients free survival by clinical and biological subgroups in the
were stratified into the low-risk group and four cohorts. The DMGN was significantly associated
71 (45%) patients were stratified into the high-risk group; with distant metastasis-free survival in all four cohorts
patients with high-risk scores had shorter distant (figure 3). Analysis using different categorisations for
metastasis-free survival (HR 3·62, 95% CI 1·43–9·19; N stage (N3 vs N0–2) and T stage (T4 vs T1–3) showed
p=0·0037; figure 2D), disease-free survival (HR 3·10, similar results to the original dichotomisations of N stage
1·48–6·47; p=0·0016; appendix p 25), and overall survival (N2–3 vs N0–1) and T stage (T3–4 vs T1–2; appendix p 27).
(HR 2·45, 1·19–5·02; p=0·012; appendix p 26). 5-year After multivariable adjustment by clinicopathological
distant metastasis-free survival, disease-free survival, variables and several serum markers, the DMGN
and overall survival in each risk group and the number remained a strong independent prognostic factor for
of patients who had an event for each risk group are distant metastasis-free survival in the Guangzhou
listed in the appendix (pp 13, 14). training cohort (HR 4·51, 95% CI 2·72–7·47; p<0·0001;

A Combined Guangzhou internal cohort: low-risk group B Combined Guangzhou internal cohort: high-risk group
100
Distant metastasis-free survival (%)

75

50

25 Concurrent chemotherapy
No concurrent chemotherapy
HR 0·40 (95% CI 0·19–0·83) HR 1·03 (95% CI 0·71–1·50)
p=0·011 p=0·876
0
0 12 24 36 48 60 72 84 96 108 120 0 12 24 36 48 60 72 84 96 108 120
Number at risk
(number
censored)
Concurrent 159 (0) 154 (2) 149 (3) 140 (6) 138 (2) 127 (11) 97 (29) 60 (36) 16 (44) 3 (13) 0 (3) 167 (0) 145 (2) 123 (2) 111 (2) 100 (5) 84 (12) 59 (25) 31 (28) 13 (17) 5 (8) 0 (5)
chemotherapy
No concurrent 153 (0) 145 (4) 136 (1) 125 (5) 115 (7) 105 (8) 88 (17) 61 (27) 29 (32) 4 (25) 0 (4) 135 (0) 117 (1) 105 (1) 95 (2) 84 (5) 75 (7) 61 (11) 39 (21) 19 (18) 3 (16) 0 (3)
chemotherapy

C Combined external cohort: low-risk group D Combined external cohort: high-risk group
100
Distant metastasis-free survival (%)

75

50

25

HR 0·38 (95% CI 0·16–0·92) HR 0·95 (95% CI 0·51–1·77)


p=0·026 p=0·871
0
0 12 24 36 48 60 72 84 96 108 0 12 24 36 48 60 72 84 96 108
Time since treatment (months) Time since treatment (months)
Number at risk
(number
censored)
Concurrent 110 (0) 107 (2) 102 (2) 100 (1) 80 (19) 54 (24) 26 (28) 17 (9) 6 (11) 0 (6) 94 (0) 88 (0) 74 (2) 61 (6) 48 (9) 29 (19) 10 (19) 0 (10) 0 (0) 0 (0)
chemotherapy
No concurrent 73 (0) 65 (1) 62 (3) 58 (0) 49 (7) 46 (3) 36 (10) 19 (17) 4 (15) 0 (4) 46 (0) 43 (0) 36 (1) 31 (1) 28 (1) 26 (2) 16 (10) 10 (6) 0 (10) 0 (0)
chemotherapy

Figure 4: Kaplan-Meier curves of distant metastasis-free survival according to low-risk or high-risk scores stratified by administration of concurrent chemotherapy
(A) DMGN-defined low-risk patients in the combined Guangzhou internal cohort (n=312), (B) DMGN-defined high-risk patients in the combined Guangzhou internal
cohort (n=302), (C) DMGN-defined low-risk patients in the combined external cohort (n=183), and (D) DMGN-defined high-risk patients in the combined external
cohort (n=140). We calculated p values using the unadjusted log-rank test and hazard ratios using a univariate Cox regression analysis. DMGN=distant metastasis
gene signature for locoregionally advanced nasopharyngeal carcinoma. HR=hazard ratio.

8 www.thelancet.com/oncology Published online February 7, 2018 http://dx.doi.org/10.1016/S1470-2045(18)30080-9


Articles

appendix p 15), as well as in the Guangzhou internal


validation cohort (HR 3·32, 1·74–6·33; p=0·00028; A
0 30 60 90
Points
appendix p 16) and the two external cohorts (Guilin:
HR 2·48, 1·31–4·71; p=0·0055; and Foshan: HR 4·88, High risk
DMGN
1·85–12·8; p=0·0014; appendix pp 17, 18). Low risk Male
We analysed whether the DMGN could be used to Sex
Female
predict the efficacy of concurrent chemotherapy in N2–3
N stage
locoregionally advanced nasopharyngeal carcinoma. 326 N0–1
≥245
(53%) of 614 patients in the combined Guangzhou cohort LDH (U/L)
received concurrent chemoradiation. When the Kaplan- <245 ≥8·2
Meier survival analysis was stratified with the DMGN, C-reactive protein (mg/L)
<8·2
treatment with concurrent chemotherapy compared with
Total points
no concurrent chemotherapy was associated with 0 50 100 150 200 250 300
improved distant metastasis-free survival (HR 0·40, 95% 3-year DMFS
CI 0·19–0·83; p=0·011), disease-free survival (HR 0·56, 0·95 0·90 0·80 0·60 0·40 0·20
0·34–0·94; p=0·025), and overall survival (HR 0·58, 5-year DMFS
0·95 0·90 0·80 0·60 0·40 0·20
0·35–0·97; p=0·036) in patients with low DMGN scores
but not in patients with high DMGN scores (distant B C
1·0
metastasis-free survival: HR 1·03, 0·71–1·50; p=0·876;
Actual 5-year DMFS (proportion)

figure 4; appendix pp 28, 29 shows the disease-free and 0·9


overall survival results). This finding was validated in the 0·8
combined external validation cohort, in which 204 (63%) 0·7
of 323 patients received concurrent chemotherapy
0·6
(figure 4; appendix pp 28, 29). The 5-year distant
0·5
metastasis-free survival, disease-free survival, and overall
survival in each risk group and the number of patients 0·4
0·4 0·5 0·6 0·7 0·8 0·9 1·0 0·4 0·5 0·6 0·7 0·8 0·9 1·0
who had an event for each risk group are listed in the Nomogram-predicted probability of 5-year DMFS Nomogram-predicted probability of 5-year DMFS
appendix (pp 13, 14). in the Guangzhou training cohort in the Guangzhou internal validation cohort
We did a multivariate analysis of distant metastasis- D E
free survival (appendix p 15) to generate nomogram A to 1·0
Actual 5-year DMFS (proportion)

predict distant metastasis-free survival in the training 0·9


cohort (figure 5A). The predictors included DMGN, sex,
0·8
N stage, and serum LDH and C-reactive protein
0·7
concentrations. Among these, DMGN had the highest
C-index (appendix p 19). The calibration plots for the 0·6

5-year distant metastasis-free survival were predicted 0·5


well in the Guangzhou training cohort (C-index 0·727, 0·4
95% CI 0·679–0·775), the Guangzhou internal 0·4 0·5 0·6 0·7 0·8 0·9 1·0 0·4 0·5 0·6 0·7 0·8 0·9 1·0
Nomogram-predicted probability of 5-year DMFS Nomogram-predicted probability of 5-year DMFS
validation cohort (0·725, 0·654–0·796), the Guilin in the Guilin external validation cohort in the Foshan external validation cohort
external validation cohort (0·693, 0·617–0·769), and the
Foshan validation cohort (0·739, 0·639–0·839; Figure 5: Nomogram A to predict the risk of distant metastasis in locoregionally advanced nasopharyngeal
figure 5B–E). ROC analysis to compare the sensitivity carcinoma
(A) Nomogram A to predict distant metastasis-free survival. Calibration curves of the nomogram to predict distant
and specificity of the nomogram comprising the DMGN metastasis-free survival at 5 years in (B) the Guangzhou training cohort, (C) the Guangzhou internal validation
signature with clinicopathological risk factors showed cohort, (D) the Guilin external validation cohort , and (E) the Foshan external validation cohort. The actual distant
that distant metastasis-free survival was more accurately metastasis-free survival is plotted on the y-axis; nomogram predicted probability is plotted on the x-axis.
predicted by the nomogram than by the risk factors in DMGN=distant metastasis gene signature for locoregionally advanced nasopharyngeal carcinoma. LDH=serum
lactate dehydrogenase. DMFS=distant metastasis-free survival.
all cohorts (appendix p 30).
Since plasma EBV DNA concentration has been shown
to be a useful prognostic biomarker in nasopharyngeal using these five risk factors plus the DMGN using the
carcinoma, we did a subgroup analysis to explore 247 patients in the Guangzhou internal cohort (appendix
whether or not it enhances the value of our present p 31). Nomogram C (C-index 0·748, 95% CI 0·680–0·816)
model. Pre-treatment EBV DNA data were available for had better accuracy for predicting distant metastasis-free
247 patients in the Guangzhou internal cohort and survival than nomogram B (0·686, 0·619–0·753;
122 patients in the Foshan external cohort (appendix appendix p 20), which was confirmed using ROC
p 21). First, we developed nomogram B using plasma analysis (p=0·037; appendix p 32). These findings were
EBV DNA status, sex, N stage, serum LDH, and validated in the 122 patients from the Foshan external
C-reactive protein, and then built a final nomogram C cohort (appendix pp 20, 32).

www.thelancet.com/oncology Published online February 7, 2018 http://dx.doi.org/10.1016/S1470-2045(18)30080-9 9


Articles

Discussion it is therefore a suitable platform for a gene expression-


In this multicentre, retrospective cohort study, we based clinical test.13,28
developed and validated a novel prognostic tool based on In this study, we showed that concurrent chemotherapy
the expression of 13 genes that compared with clinical versus no concurrent chemotherapy provides a survival
risk factors improves the ability to predict distant benefit to patients classified as low risk by our DMGN
metastasis in patients with locoregionally advanced tool, but not in those classified as high risk. These results
nasopharyngeal carcinoma. Our results showed that the suggest that low-risk patients could be cured by
DMGN developed in this study categorised patients into concurrent chemotherapy, whereas high-risk patients are
high-risk and low-risk groups of patients who had candidates for more aggressive therapeutic strategies to
significantly different distant metastasis-free survival. prevent tumour metastasis. Concurrent chemotherapy,
Furthermore, our DMGN classifier was significantly due to its additive or synergistic effect with radiotherapy,
better than other clinicopathological risk factors at provides a survival benefit mainly by improving
predicting distant metastasis-free survival in patients locoregional control but not metastasis.29 It is possible
with locoregionally advanced nasopharyngeal carcinoma. that concurrent chemotherapy does not effectively
We also built a nomogram based on the DMGN and other eradicate micrometastases in the high-risk group.
variables that predicts individual risk of distant metastasis, Cancer is a heterogeneous disease, and exploring the
which was strengthened by adding EBV DNA status. dysregulated genes involved in carcinogenesis and
Advances in molecular biology have resulted in the development might help to improve prognostic and
generation of large amounts of data that have been used therapeutic strategies. In the current study, we identified
to construct multigene profiles, which can be used for a group of 13 genes that effectively predict nasopharyngeal
risk stratification and to guide strategies for chemotherapy carcinoma distant metastasis and concurrent chemo­
treatment in various types of cancers.10–13 Although a few sensitivity. Among these genes, FNDC3B and CLASP1
gene-expression profiling studies have been done to were previously shown to regulate cell motility and to
identify genes that are differentially expressed between thereby influence metastatic ability; and ANXA1 and
nasopharyngeal carcinoma and non-cancerous tissues, CXCL10 were found to be involved in immune responses
differences in expression profiles between patients with and the NF-κB signalling pathway, which influences
different prognoses are largely unexplored.14–16 Moreover, tumour apoptosis, angiogenesis, and invasiveness.30–35
while previous gene-expression profiling studies have Clinically promising results have been achieved by
yielded large numbers of markers, they did not subject inhibiting VEGF and using immunotherapies, such as
them to large-scale, independent validation. In this study, adoptive T-cell transfer, in patients with recurrent
we used microarray analysis to compare global gene- nasopharyngeal carcinoma.36,37 Therefore, our DMGN
expression profiles between tissue samples obtained could potentially be used as a predictive tool in
from paired locoregionally advanced nasopharyngeal personalised therapy, and might offer potential targets
carcinoma tumours with and without distant metastasis for therapeutic intervention in the clinical management
after radical treatment. Digital expression profiling was of nasopharyngeal carcinoma.
then used to generate a DMGN classifier that predicted We built a nomogram to predict an individual’s risk of
the risk of distant metastasis in locoregionally advanced distant metastasis. Despite the fact that such methods
nasopharyngeal carcinoma, and the results were generally use traditional prognostic factors, such as N stage
validated in an internal validation cohort and two external and sex, we propose that including our DMGN reflects the
independent validation cohorts. Importantly, this biological heterogeneity of these tumours, whereas blood
classifier was applicable to patients receiving either 2D parameters, such as serum C-reactive protein and LDH
radiotherapy (Guilin cohort) or intensity-modulated concentrations, provide insights into a patient’s systemic
radiation therapy (Foshan cohort). Considering that inflammatory status and elevated concentrations of these
PET–CT is able to discover 6–12% more cases of distant are associated with inferior survival in nasopharyngeal
metastasis than staging work-up with a conventional carcinoma.7,22 The performance of the nomogram was
abdominal B ultrasound, chest radiography, and bone verified in all validation cohorts. Moreover, the addition of
scan,25,26 DMGN remained an independent prognostic EBV DNA status to this nomogram improved predictive
factor after inclusion of PET–CT as a covariate in accuracy, possibly because EBV DNA concentration
multivariable analysis. Additionally, the fact that our reflects tumour burden. Thus, our nomogram might
approach uses the US Food and Drug Administration- provide a simple and accurate method for predicting
approved NanoString platform makes it easy to prognoses in locoregionally advanced nasopharyngeal
implement in clinical practice. Moreover, the mRNA carcinoma.
platform is Clinical Laboratory Improvement Our study had several limitations. First, our data were
Amendments certified, and was used to implement obtained in an endemic area in China where the
PAM50 for breast cancer subtyping.27 This technology nasopharyngeal cancer pathological subtype is mainly
has been shown to be robust and reliable for quantifying undifferentiated non-keratinising carcinoma; the
RNA species that were extracted from FFPE tissues, and pathological subtype and distribution of clinical

10 www.thelancet.com/oncology Published online February 7, 2018 http://dx.doi.org/10.1016/S1470-2045(18)30080-9


Articles

characteristics might be different in other areas. Second, 5 Hui EP, Leung SF, Au JS, et al. Lung metastasis alone in
nasopharyngeal carcinoma: a relatively favorable prognostic group.
the biological mechanisms by which many candidate A study by the Hong Kong Nasopharyngeal Carcinoma Study
markers that have been included in the DMGN, such as Group. Cancer 2004; 101: 300–06.
POLR2M and YBX3, contribute to nasopharyngeal 6 Lin JC, Chen KY, Wang WY, et al. Detection of Epstein-Barr virus
carcinoma metastasis remain unknown, and further DNA in the peripheral-blood cells of patients with nasopharyngeal
carcinoma: relationship to distant metastasis and survival.
investigations into their functions might provide novel J Clin Oncol 2001; 19: 2607–15.
targets and treatment strategies. Moreover, our DMGN 7 Zhou GQ, Tang LL, Mao YP, et al. Baseline serum lactate
and nomogram require further validation in prospective dehydrogenase levels for patients treated with intensity-modulated
radiotherapy for nasopharyngeal carcinoma: a predictor of poor
studies and multicentre clinical trials; we are proceeding prognosis and subsequent liver metastasis.
with a prospective, multicentre study (NCT03025854). Int J Radiat Oncol Biol Phsy 2012; 82: e359–65.
To our knowledge, this is the first study to include 8 Liu N, Chen NY, Cui RX, et al. Prognostic value of a microRNA
signature in nasopharyngeal carcinoma: a microRNA expression
such a large sample size that has evaluated the global analysis. Lancet Oncol 2012; 13: 633–41.
gene-expression profiles in locoregionally advanced 9 Kim KY, Le Q, Yom SS, et al. Clinical utility of Epstein-Barr virus
naso­pharyngeal carcinoma tumours with different DNA testing in the treatment of nasopharyngeal carcinoma
patients. Int J Radiat Oncol Biol Phys 2017; 98: 996–1001.
prognoses. Our findings show that the DMGN
10 Cardoso F, van’t Veer LJ, Bogaerts J, et al. 70-gene signature as an
prognostic tool can effectively classify patients with aid to treatment decisions in early-stage breast cancer. N Engl J Med
locoregionally advanced nasopharyngeal carcinoma into 2016; 375: 717–29.
groups with different risks of distant metastasis, thereby 11 Zhao SG, Chang SL, Spratt DE, et al. Development and validation
of a 24-gene predictor of response to postoperative radiotherapy in
adding prognostic value to the traditional clinico­ prostate cancer: a matched, retrospective analysis. Lancet Oncol
pathological risk factors used to assess patient 2016; 17: 1612–20.
prognoses. Moreover, in our study, we show that the 12 Rini B, Goddard A, Knezevic D, et al. A 16-gene assay to predict
recurrence after surgery in localised renal cell carcinoma:
DMGN classifier might be a useful predictive tool for development and validation studies. Lancet Oncol 2015; 16: 676–85.
identifying patients who would benefit from concurrent 13 Scott DW, Chan FC, Hong F, et al. Gene expression-based model
chemotherapy. A nomogram comprising DMGN might using formalin-fixed paraffin-embedded biopsies predicts overall
survival in advanced-stage classical Hodgkin lymphoma.
help clinicians in directing personalised therapeutic J Clin Oncol 2013; 31: 692–700.
regimen selection for patients with locoregionally 14 Shi W, Bastianutto C, Li A, et al. Multiple dysregulated pathways in
advanced nasopharyngeal carcinoma. nasopharyngeal carcinoma revealed by gene expression profiling.
Int J Cancer 2006; 119: 2467–75.
Contributors 15 Zeng Z, Zhou Y, Xiong W, et al. Analysis of gene expression
JM and NL designed the study. X-RT, NL, Y-QL, S-BL, WJ, FL, Q-MH, identifies candidate molecular markers in nasopharyngeal
X-JY, and YZ collected the data. JM, NL, X-RT, W-XG, Y-QL, Q-MH, L-LT, carcinoma using microdissection and cDNA microarray.
and Y-PM analysed and interpreted the data. X-RT, JM, and NL wrote the J Cancer Res Clin Oncol 2007; 133: 71–81.
manuscript. Y-QL, XW, JZ, Y-QW, P-PZ, YS, J-PY, JZ, L-ZL, and LL 16 Wang S, Li X, Li ZG, et al. Gene expression profile changes and
revised the manuscript. X-RT, W-XG, and NL did the statistical analysis. possible molecular subtypes in differentiated-type nonkeratinizing
All authors reviewed the manuscript and approved the final version. nasopharyngeal carcinoma. Int J Cancer 2011; 128: 753–62.
17 Amin MB, Edge S, Greene F, et al. AJCC cancer staging manual,
Declaration of interests
8th edn. New York, NY: Springer, 2016.
We declare no competing interests.
18 Zhang JX, Song W, Chen ZH, et al. Prognostic and predictive value
Acknowledgments of a microRNA signature in stage II colon cancer: a microRNA
We thank Hanqi Yin (Shanghai Biotechnology Corporation, Shanghai, expression analysis. Lancet Oncol 2013; 14: 1295–306.
China) for statistical consultation, and Yiducloud (Beijing) Technology 19 Nault JC, De Reyniès A, Villanueva A, et al. A hepatocellular
Ltd for supporting part of the data extraction and processing. This study carcinoma 5-gene score associated with survival of patients after
was supported by grants from the National Natural Science Foundation liver resection. Gastroenterology 2013; 145: 176–87.
of China (81230056, 81402516), the National Science & Technology Pillar 20 Guo P, Luo Y, Mai G, et al. Gene expression profile based
Program during the Twelfth Five-year Plan Period (2014BAI09B10), classification models of psoriasis. Genomics 2014; 103: 48–55.
Natural Science Foundation of Guang Dong Province (2017A030312003), 21 Camp RL, Dolled-Filhart M, Rimm DL. X-tile: a new bio-informatics
the National Key Research and Development Program of China tool for biomarker assessment and outcome-based cut-point
(2016YFC0902000), the Innovation Team Development Plan of the optimization. Clin Cancer Res 2004; 10: 7252–59.
Ministry of Education (IRT_17R110), Health & Medical Collaborative 22 Xia WX, Zhang HB, Shi JL, et al. A prognostic model predicts the
Innovation Project of Guangzhou City, China (201400000001), and the risk of distant metastasis and death for patients with nasopharyngeal
carcinoma based on pre-treatment serum C-reactive protein and
Program of Introducing Talents of Discipline to Universities (B14035).
N-classification. Eur J Cancer 2013; 49: 2152–60.
References 23 Tang LQ, Li CF, Li J, et al. Establishment and validation of
1 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. prognostic nomograms for endemic nasopharyngeal carcinoma.
Global cancer statistics, 2012. CA Cancer J Clin 2015; 65: 87–108. J Natl Cancer Inst 2016; 108: pii: djv291.
2 Pan JJ, Ng WT, Zong JF, et al. Prognostic nomogram for refining 24 Heagerty PJ, Lumley T, Pepe MS. Time-dependent ROC curves for
the prognostication of the proposed 8th edition of the AJCC/UICC censored survival data and a diagnostic marker. Biometrics 2000;
staging system for nasopharyngeal cancer in the era of 56: 337–44.
intensity-modulated radiotherapy. Cancer 2016; 122: 3307–15. 25 Chang JT, Chan SC, Yen TC, et al. Nasopharyngeal carcinoma
3 National Comprehensive Cancer Network guidelines. 2017. staging by (18)F-fluorodeoxyglucose positron emission tomography.
http://www.nccn.org/professionals/physician_gls/pdf Int J Radiat Oncol Biol Phys 2005; 62: 501–07.
(accessed Aug 23, 2017). 26 Tang LQ, Chen QY, Fan W, et al. Prospective study of tailoring
4 Lai SZ, Li WF, Chen L, et al. How does intensity-modulated whole-body dual-modality [18F]fluorodeoxyglucose positron emission
radiotherapy versus conventional two-dimensional radiotherapy tomography/computed tomography with plasma Epstein-Barr virus
influence the treatment results in nasopharyngeal carcinoma DNA for detecting distant metastasis in endemic nasopharyngeal
patients? Int J Radiat Oncol Biol Phys 2011; 80: 661–68. carcinoma at initial staging. J Clin Oncol 2013; 31: 2861–69.

www.thelancet.com/oncology Published online February 7, 2018 http://dx.doi.org/10.1016/S1470-2045(18)30080-9 11


Articles

27 Sestak I, Cuzick J, Dowsett M, et al. Prediction of late distant 32 Zhang Z, Huang L, Zhao W, et al. Annexin 1 induced by
recurrence after 5 years of endocrine treatment: a combined anti-inflammatory drugs binds to NF-κB and inhibits its activation:
analysis of patients from the Austrian Breast and Colorectal Cancer anticancer effects in vitro and in vivo. Cancer Res 2010; 70: 2379–88.
Study Group 8 and Arimidex, Tamoxifen Alone or in Combination 33 Vacchelli E, Enot DP, Pietrocola F, et al. Impact of pattern
randomized trials using the PAM50 risk of recurrence score. recognition receptors on the prognosis of breast cancer patients
J Clin Oncol 2015; 33: 916–22. undergoing adjuvant chemotherapy. Cancer Res 2016; 76: 3122–26.
28 Ramaswamy V, Remke M, Bouffet E, et al. Recurrence patterns 34 Brownell J, Polyak SJ. Molecular pathways: hepatitis C virus,
across medulloblastoma subgroups: an integrated clinical and CXCL10, and the inflammatory road to liver cancer. Clin Cancer Res
molecular analysis. Lancet Oncol 2013; 14: 1200–07. 2013; 19: 1347–52.
29 Lin JC, Jan JS, Hsu CY, et al. Phase III study of concurrent 35 Harris DP, Bandyopadhyay S, Maxwell TJ, et al. Tumor necrosis
chemotherapy versus radiotherapy alone for advanced factor (TNF)-α induction of CXCL10 in endothelial cells requires
nasopharyngeal carcinoma: positive effect on overall and protein arginine methyltransferase 5 (PRMT5)-mediated nuclear
progression-free survival. J Clin Oncol 2003; 21: 631–37. factor (NF)-κB p65 methylation. J Biol Chem 2014; 289: 15328–39.
30 Zhang X, Liu S, Hu T, et al. Up-regulated microRNA-143 36 Hui EP, Ma BBY, King AD, et al. Hemorrhagic complications in a
transcribed by nuclear factor kappa B enhances hepatocarcinoma phase II study of sunitinib in patients of nasopharyngeal carcinoma
metastasis by repressing fibronectin expression. Hepatology 2009; who has previously received high-dose radiation. Ann Oncol 2011;
50: 490–99. 22: 1280–87.
31 Bouchet BP, Noordstra I, van Amersfoort M, et al. 37 Chia WK, Teo M, Wang WW, et al. Adoptive T-cell transfer and
Mesenchymal cell invasion requires cooperative regulation of chemotherapy in the first-line treatment of metastatic and/or locally
persistent microtubule growth by SLAIN2 and CLASP1. Dev Cell recurrent nasopharyngeal carcinoma. Mol Ther 2014; 22: 132–39.
2016; 39: 708–23.

12 www.thelancet.com/oncology Published online February 7, 2018 http://dx.doi.org/10.1016/S1470-2045(18)30080-9

You might also like