Research Article

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Hindawi

Journal of Oncology
Volume 2019, Article ID 3865279, 10 pages
https://doi.org/10.1155/2019/3865279

Research Article
Bioinformatic Analysis Reveals an Immune/
Inflammatory-Related Risk Signature for Oral Cavity Squamous
Cell Carcinoma

Shuang Bai ,1,2 Ying-Bin Yan,1 Wei Chen,1 Ping Zhang,1 Tong-Mei Zhang,1
Yuan-Yuan Tian,1 and Hao Liu 1
1
Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Nankai University, Tianjin Stomatological Hospital,
No. 75, Dagu Road, Heping District, Tianjin 300041, China
2
Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, No. 22,
Zhongguancun South Avenue, Haidian District, Beijing 100081, China

Correspondence should be addressed to Shuang Bai; cmubaishuang@hotmail.com and Hao Liu; tjshhaoliu@hotmail.com

Received 1 August 2019; Revised 6 October 2019; Accepted 5 November 2019; Published 13 December 2019

Guest Editor: Qigen Fang

Copyright © 2019 Shuang Bai et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
High-throughput gene expression profiling has recently emerged as a promising technique that provides insight into cancer
subtype classification and improved prediction of prognoses. Immune/inflammatory-related mRNAs may potentially enrich
genes to allow researchers to better illustrate cancer microenvironments. Oral cavity squamous cell carcinoma (OC-SCC) exhibits
high morbidity and poor prognosis compared to that of other types of head and neck squamous cell carcinoma (HNSCC), and
these differences may be partially due to differences within the tumor microenvironments. Based on this, we designed an immune-
related signature to improve the prognostic prediction of OC-SCC. A cohort of 314 OC-SCC samples possessing whole genome
expression data that were sourced from The Cancer Genome Atlas (TCGA) database was included for discovery. The GSE41613
database was used for validation. A risk score was established using immune/inflammatory signatures acquired from the training
dataset. Principal components analysis, GO analysis, and gene set enrichment analysis were used to explore the bioinformatic
implications. When grouped by the dichotomized risk score based on the signature, this classifier could successfully discriminate
patients with distinct prognoses within the training and validation cohorts (P < 0.05 in both cohorts) and within different
clinicopathological subgroups. Similar somatic mutation patterns were observed between high and low risk score groups, and
different copy number variation patterns were also identified. Further bioinformatic analyses suggested that the lower risk score
group was significantly correlated with immune/inflammatory-related biological processes, while the higher risk score group was
highly associated with cell cycle-related processes. The analysis indicated that the risk score was a robust predictor of patient
survival, and its functional annotation was well established. Therefore, this bioinformatic-based immune-related signature
suggested that the microenvironment of OC-SCC could distinguish among patients with different underlying biological processes
and clinical outcomes, and the use of this signature may shed light on future OC-SCC classification and therapeutic design.

1. Introduction but do not overlap. There are differences in the associated


mortality, etiology, risk factors, and even the biomarkers of
Oral cavity squamous cell carcinoma (OC-SCC) is the most squamous cell carcinomas located within these two major
common malignancy of the head and neck region (excluding sites.
nonmelanoma skin cancer). There has also been a recent Many risk factors can contribute to OC-SCC. Tobacco is
dramatic rise in the incidence of oropharyngeal squamous classified as a group 1 carcinogen for OC-SCC. Currently,
cell carcinoma (OP-SCC) [1]. Anatomically, the mouth and tobacco smokers exhibit a 3.43 fold relative risk for OC-SCC
oropharynx are separate areas that are adjacent to each other compared to that of nonsmokers [2], and they possess a fully
2 Journal of Oncology

adjusted HR of 1.7 [3]. Alcohol has been established as an were extracted from the Molecular Signatures Database v6.1
independent risk factor, and studies of nonsmokers have (http://http://software.broadinstitute.org/gsea/msigdb/
demonstrated a strong correlation and dose-response re- index.jsp) and were combined to integrate the immune/
lationship between alcohol consumption and OC-SCC [4]. inflammatory-related gene set.
Additionally, the combined effect of smoking and alcohol
consumption was greater if the relationship was multipli-
2.2. Statistical Analysis. Overall survival time (OS) was
cative [5]. HPV, as a major etiological factor, contributes
defined as the interval from the date of diagnosis until
disproportionally to the formation and prognosis of squa-
death or until the last follow-up. The prognostic value for
mous cell carcinoma formation at different sites within the
patients possessing high or low expression of a certain gene
head and neck region [6]. Specifically, the high-risk genotype
or score (higher or lower than the median value) was
HPV-16 accounts for the vast majority (about 90% to 95%)
calculated using the Kaplan–Meier method with the two-
of HPV-positive OP-SCC, while the HPV type is more
sided log-rank test by package “survival” of R. Univariate
variable in OC-SCC [7, 8]. Additionally, a far more favorable
and multivariate COX regression analysis was also per-
outcome exists for HPV positive compared to that for HPV-
formed using package “survival” of R. Chi-squared test and
negative OP-SCC [9].
The above three major risk factors may contribute to Fisher’s exact test were used to compare the frequencies
carcinogenesis through different biological processes or between groups. A two-tailed Student’s t-test was per-
formed to compare two groups of numerical values.
pathways [10–12]; however, the dysregulation of immune or
Analysis of variance (ANOVA) was used to analyze the
inflammatory responses is mutually shared among the
differences among group means. The median absolute
carcinogenic mechanisms. Moderate alcohol consumption
deviation (MAD) calculated in R. Pearson correlation
or alcohol abuse can suppress multiple arms of the immune
analysis was used to evaluate the association between two
response [13, 14]. Cigarette smoke exposure significantly
variables and was calculated by R function “cor.test.” The
affects the immune system, impairing the ability of the host
statistical analysis was performed using the software of R
to produce appropriate immune and inflammatory re-
version 3.4 for Windows. The statistical significance was
sponses, ultimately leading to smoking-related pathologies
established at the level of P < 0.05.
[15]. These concepts are consistent with the notion that head
and neck cancer is an intrinsically immune-suppressing
disease [16]. For HPV-positive SCC, more intense PD-L1+, 2.3. Bioinformatic Analysis. The R “Limma” package, a
CD4+, and CD8+ T-cell infiltration correlates with a better package that can perform the differential expression analyses
outcome [17]. Increasing evidence supports the idea that of RNA sequencing (RNA-seq) data [23], was used to
evoked immune or inflammatory responses may elicit an- identify differentially expressed genes (DEGs) based on a
titumor effects concerning OC-SCC development [18, 19]. threshold of false discovery rate (FDR) of less than 0.05. The
Our current study aimed to develop a key gene signature packages “gaia,” “maftool,” and “circlize” were used to
that is representative of immune and inflammatory re- generate mutation and CNV plots [24]. The R “TCGAbio-
sponses that could be correlated with patient prognosis, and links” package was employed to investigate relevant bi-
we incorporated a bioinformatics-based approach associated ological implications [25]. The biological phenotype was
with clinical covariates to achieve our aim. Following this further verified by gene set enrichment analysis (GSEA) [22].
principle, we performed a combined analysis to identify a Normalized enrichment score (NES) and false discovery rate
robust gene signature, and we established a risk score sys- were used to determine the statistical significance. The R
tem. Further bioinformatic analyses revealed that the risk “ESTIMATE” package that was fitted for our data was used
score exhibited excellent prognostic value for stratifying to calculate ImmuneScore, StromalScore, and tumor purity
patients irrespective of mutation or copy number variation [26].
(CNV) patterns, and this risk score was highly associated
with cell-cycle processes. 3. Results
2. Materials and Methods 3.1. Different Immune/Inflammatory Phenotypes of OC-SCC
Tumor. The gene expression and clinical data for 314 pa-
2.1. Datasets. Whole genome mRNA expression RNA-seq tients were obtained from the TCGA database (Table S1).
(20101 genes) data, somatic mutation data, copy number Previous research defined four RNA subtypes of the TCGA
variation data, and all corresponding clinical information cohort, including atypical, basal, classical, and mesenchymal
from the TCGA-HNSC dataset [20] (http://cancergenome. [20]. The combined gene set that was representative of
nih.gov/) were downloaded for use as the training cohort. immune/inflammatory response (1224 genes) was used to
The following dataset was obtained for validation: GSE41613 illustrate the immune microenvironment of the OC-SCC
(23520 genes) [21] (http://www.ncbi.nlm.nih.gov/geo/ tumor. Principal components analysis based on the 1224
query/acc.cgi?acc�GSE41613). The training dataset com- genes revealed a different distribution pattern regarding OC-
prised 314 OC-SCC patients while the validation dataset SCC tumor subtypes. A mutually exclusive pattern was
comprised 97 OC-SCC patients. The patient characteristics observed within the mesenchymal, basal, and classical
are summarised in Table S1. Two gene sets (immune re- subtypes, while the atypical subtype lacked a clear distri-
sponse, M14329 and inflammatory response, M13657) [22] bution pattern (Figure 1(a)). As clinical differences between
Journal of Oncology 3

–0.08
–0.1
0 –0.075
0.1
–0.07
0.2

–0.1

PC3
0

0.1

0.2

PC
2
PC

Basal 1 Atypical
Mesenchymal Classical
(a)

Go_inflammatory_response (454 genes) The cancer genome atlas dataset (20101 genes) Go_immune_response (1100 genes)

1224 common genes

Heterogeneity analysis (MAD > 1.0)

668 genes

Univariate Cox regression (P < 0.01)

β-value of
18 genes
each gene

Risk score = expression value of each gene × β-value of each gene

(b)

Figure 1: Different immune/inflammatory patterns of OC-SCC subtypes and the workflow of the signature establishment. (a) Principal
components analysis of immune/inflammatory genes among four subtypes of OC-SCC. (b) Workflow of the signature establishment.

the four tumor subtypes were previously established, we expressed (MAD ≤ 1) were excluded from further analysis.
sought to construct a gene signature to further explore the Univariate Cox regression analysis was used to explore the
immune phenotype of OC-SCC tumors. prognostic value of the resulting genes. Eighteen genes
(CD27, CD79B, CMA1, CCR4, CCR7, CNR2, CTLA4,
CTSG, GZMM, IL16, MASP1, SAA1, CCL11, TNFAIP3,
3.2. Identification of an Immune/Inflammatory Signature for BATF, IL19, PGLYRP4, and TREML1) were identified to be
Prognosis Prediction in OC-SCC. Taking into account the associated with prognosis in OC-SCC patients (Figures 1(b)
differential distribution of immune/inflammatory genes and 2(a)).
among OC-SCC subtypes, we sought to identify the gene or Next, the risk score method was employed based on
group of genes that were of significant prognostic value. As gene expression levels (coefficient of each gene was the
the PCA exhibited a heterogeneous expression pattern beta value in the univariate Cox regression model of each
among OC-SCC patients, genes that were homogeneously gene; Table S2), and the associations between TCGA
4 Journal of Oncology

Color key TCGA OSCC


Training cohort 100

Overall survival (%)


–2 0 2
Row Z-score
Risk score 50 P = 1.13e – 05
Tumor stage
RNA subtype
Methylation
Gender 0
SAA1 0 2000 4000 6000
TNFAIP3
CCR7 Low risk (n = 157)
CD79B High risk (n = 157)
PGLYRP4
CD27 (b)
BATF
CCL11
CTLA4
IL19 GSE41613 HPV negative
CCR4
TREML1 100

Overall survival (%)


MASP1
CMA1
GZMM P = 0.0162
IL16
CNR2 50
CTSG
Tumor stage RNA subtype Methylation
III/IV AT Hypermethylated
0
I/II BA Hypomethylated 0 1000 2000 3000
Gender CL CpG hypermet
Male ME Normal-like Low risk (n = 48)
Female High risk (n = 49)

(a) (c)

Figure 2: Overview of the eighteen-gene-based risk score and its prognostic value across different cohorts. (a) Heatmap depicting the gene
expression values of the eighteen genes comprising the signature of the training cohort. Columns representing each sample that were sorted
by increasing value of the risk score. Rows representing the expression value of each gene. (b) Kaplan–Meier survival analyses based on the
median cutoff of the risk score within the training dataset. (c) Kaplan–Meier survival analyses based on the median cutoff of the risk scores
within the validation dataset.

results and two functional gene sets were explored. A group within each subgroup to query the prognostic value. A
strong correlation was identified between the risk score nearly universal result was achieved among most of the
and the ssGSEA score in the immune/inflammatory re- subgroups (Figure S2(B–J)), demonstrating that an elevated
sponse gene set (R � − 0.693, P < 0.0001; Figure S1A). The risk score was strongly correlated with poor prognosis and
resulting 18 genes were also mutually correlated, and this vice versa. Similar results were observed for the validation
supported the integrity of the signature genes (Figure S1B GSE41613 cohort (Figure S3). Univariate and multivariate
and S1C). Patients were divided into high-risk and low- Cox regression analyses also indicated that the risk score
risk groups based on the median cutoff value of the risk provided an independent prognostic factor after adjusting
score. This classifier could stratify patients according to for other clinical covariates (Table 1). Of note, tobacco use,
distinct prognosis within the training cohort (median alcohol consumption, and HPV infection, when assessed as
OS � 804 vs 2166 days; P � 1.13e − 05; Figure 2(b)) and initiating risk factors for OC-SCC, did not provide signif-
within the validation cohort (P � 0.0162; Figure 2(c)). icant HR value in either univariate or multivariate Cox
analyses. We further explored the relationship between
tobacco use, alcohol consumption, or HPV infection and the
3.3. Distribution and Prognostic Value of the Risk Score among risk score, and we found that there were no significant
Subgroups of OC-SCC. The patients within the training differences.
cohort were further stratified based on several clinico-
pathological factors, including age, gender, stage, RNA
subtype, and methylation subtype. We found that in patients 3.4. Mutation and CNV Patterns of Different Subgroups of the
classified at a higher stage, the classical and hypomethylated Risk Score. To further investigate the impact of the risk score
subtypes exhibited a higher risk score (Figure S2A). Based on at the DNA level, TCGA cases possessing available somatic
the median cutoff value of risk score in the training cohort, mutation and copy number variation (CNV) information
the patients were divided into either a high- or low-risk were analyzed. Based on the increasing risk score, cases were,
Journal of Oncology 5

Table 1: COX regression analysis of the risk score and other and a higher risk score was highly correlated with cell-cycle-
characteristics in the TCGA OC-SCC cohort. related processes (Figure 4(d)).
Univariate Multivariate
Cox Cox
Variables regression regression 3.6. Association between the Risk Score and Tumor Purity.
It is established that OC-SCC tissues contain abundant
HR P HR P
nontumor cells within their microenvironment. Thus, tumor
Risk score (high vs low) 2.09 1.7e − 05 1.99 0.0002
purity could reflect the tumor and nontumor compartments
Age (>60 vs ≤60) 1.15 0.3963 1.31 0.1580
Gender (female vs male) 1.08 0.6698 1.10 0.6357
of the OC-SCC tissue. As analyzed above, the risk score was
Tobacco history (yes vs no) 1.29 0.2099 1.27 0.2740 closely associated with immune/inflammatory responses.
Alcohol history (yes vs no) 1.07 0.6958 1.01 0.9507 Based on this, we further explored the correlation between
HPV status (positive vs negative) 0.88 0.7502 0.86 0.7138 tumor purity and the risk score. The risk score exhibited a
Stage (III/IV vs I/II) 2.23 0.0006 2.10 0.0023 high negative correlation with ImmuneScore and Stromal-
HR: hazard ratio. Score and a positive correlation with tumor purity (Fig-
ure 5). The results obtained using the validation cohort
GSE41613 were in accordance with those obtained using the
respectively, divided into four subgroups, and the most training cohort (Figure S4). These results further validated
representative subgroups (lower quantile risk score group, the idea that the risk score provided a robust predictor of
n � 78 and higher quantile risk score group, n � 78) were OC-SCC immune/inflammatory responses; however, the
selected. dichotomized median value of either of the purity-related
An analysis of the 20 most frequently mutated genes scores was unable to achieve a significant prognostic value.
within either subgroup was performed (Figure 3). Well-
known mutated genes such as TP53, a somatic mutation
detected in 60–80% of OSCC [27], were a genome guardian 4. Discussion
and played a pivotal role in regulating the cell cycle, cellular
As the most common malignancy of the head and neck, OS-
differentiation, DNA repair, and apoptosis and showed no
SCC is diagnosed using histopathological criteria and is
significant mutation difference. There was also no significant
staged using the TNM system [1]. Several studies have
difference regarding FAT1 mutation, which played a role in
provided high-resolution images of the OC-SCC molecular
regulating the migration and invasion of OSCC cells through
landscape, and these images revealed significant changes that
the localization of β-catenin [28]. Only frequent mutations
may contribute to the pathogenesis and biology of this
in NSD1 (P � 0.034) were significantly enriched in cases
disease [20]. Risk factors such as tobacco use, alcohol
with higher risk score. Subsequently, CNV data were in-
consumption, and HPV infection have been proposed as
vestigated, and our results revealed similar overall variant
initiating risk factors for OC-SCC [29–31]; however, there is
counts between OC-SCC at both lower and higher risk
little consensus on how immune/inflammatory responses
scores (mean 108.2 variants vs 111.6 variants; P � 0.8713).
could affect OS-SCC subtypes. The development of mean-
There were some differences in the gene-level CNV land-
ingful signatures to determine the immune status of patients
scape; however, the frequently deleted genomic regions were
provides an attractive therapeutic approach to this disease,
located at the 9p21.3 region encompassing the CDKN2A/
as these signatures not only promise to be powerful prog-
CDKN2B (mean deletion − 0.002 vs − 0.138, P � 0.015). The
nostic biomarkers, but if properly applied, they also stratify
7p11.2 region encompassing EGFR (mean amplification
patients to increase the likelihood of positive outcomes in
0.083 vs 0.212, P � 0.016) was frequently amplified for cases
response to immunotherapy. In our study, we demonstrated
with higher risk scores (Figure 3).
that the four subtypes of OC-SCC represent distinct im-
mune/inflammatory phenotypes, and based on this, we
3.5. High Risk Score OC-SCC Exhibited Cell-Cycle-Related established a gene signature that could stratify patients to
Gene Function While Low Risk Score Exhibited Immune/In- provide different prognoses.
flammatory Responses. To further explore the prognostic For our signature, we investigated the DNA level sce-
value, similar patterns of mutation, and the CNV of the risk nario and discovered similar mutational and CNV patterns
score, GO analysis was performed to assess the functional that were present in either high or low risk score groups. This
aspects. Pearson correlation score (R) was calculated for each result of the mutated genes suggested that it was the in-
gene within the training cohort. GO results based on 1355 flammatory responses and microenvironment that mainly
negatively correlated (R < − 0.4) genes suggested that these contribute to the different prognosis rather than well-known
genes were highly enriched in immune/inflammatory re- tumorigenesis progresses. Only frequent mutations in NSD1
sponses (Figure 4(a)). Additionally, GO results based on were significantly enriched in cases with higher risk scores. It
1632 positively correlated (R > 0.2) genes suggested that was reported that NSD1 was more mutated in laryngeal and
these genes were highly enriched in cell-cycle-related pro- pharyngeal squamous cell carcinoma (L/P-SCC) than in
cesses (Figure 4(b)). We next performed gene set enrichment OC-SCC [32], and given the role of NSD1 as a chromatin
analysis for further validation. GSEA revealed that a lower modifier, these mutations could contribute to cancer for-
risk score was associated with processes or pathways closely mation through a combination of rare germline variants and
related to immune/inflammatory responses (Figure 4(c)), somatic loss-of-heterozygosity (LOH) [33].
6 Journal of Oncology

12 10
10 8
8
6 6
4 0 10 20 30 40 50 4
2 2 0 10 20 30 40 50
0 0
69 TP53 77 TP53
35 TTN 35 TTN
29 CDKN2A 30 FAT1
23 FAT1 22 CDKN2A
18 NOTCH1 17 SYNE1
18 PIK3CA 16 LRP1B
15 PCLO 14 NOTCH1
13 MUC16 14 CASP8
13 CSMD3 14 CASMD3
12 USH2A 14 DNAH5
12 SYNE1 14 PCLO
(%)

(%)
12 CASP8 13 PIK3CA
12 FLG 12 MUC16
10 LRP1B 12 KMT2D
10 KAT4 10 FLG
10 KMT2D 10 PLEC
9 DNAH5 9 NSD1
9 HUWE1 8 FAT4
5 AHNAK 8 HRAS
5 HRAS 8 HUWE1
3 PLEC 6 AHNAK
1 NSD1 3 USH2A

Nonsense_Mutation Splice_Site Frame_Shift_Ins Nonsense_Mutation Splice_Site Frame_Shift_Ins


Missense_Mutation Multi_Hit In_Frame_Ins Missense_Mutation Multi_Hit In_Frame_Ins
Frame_Shift_Del In_Frame_Del Frame_Shift_Del In_Frame_Del

(a) (b)
14 15 16 15 16
90 M B 0 MB 90 M B 0 M 14 90MB 0
MB 9 0 MB 0 M
17
B
17 B
13 0 MB 90 M 90 M
90 M
B B 0 MB
13 90 M
B
0MB B 0M
B

B 0M 18 0M
B
18
0M B B
0M
12 90 MB 19 19
12
0M
0M B B
M
B 90
B 0 0
M B

20
M
20
M

B
0

0M
B
B

0M
11

21
M

B
11
21B
0M

M
90

B
90

0M

22
0M
22
B

B
0M

B
B

0M

0M
0M
B

B
90 M

B
10

90 M
10

X
X

90 M
90 MB
0 MB

B
0 MB
9 0 MB

0 MB
90 MB
0 MB
Y

Y
9

0M B
0 MB
0 MB

0 MB

90 M B
9 0 MB
90 MB

9 0 MB

1
1

8
8

MB

MB
0 MB

1 80

18 0
0M
B
B

B
0M
90 M

0M
90 M
B

B
7
7

B
M

M
0M

90

0M

90
B

2
2
B

0M

B
90
18 90 0M
M M 18

6
B 6 B
B
0M B 0M 0M
B 0M B
180
18 0 M
MB B B
90 MB
90 M 90 M
90 M B
B
1 80 M
B
3 5 0 MB 1 80 M
B
3
5 0 MB
1 8 0 MB 90 MB
0 MB 1 80 MB 90 M B 0 MB

4 4

Mutations CNVs Mutations CNVs


Missense_Mutation Amp Missense_Mutation Amp
Frame_Shift_Ins Del
Frame_Shift_Ins Del
Nonsense_Mutation

(c) (d)

Figure 3: Different mutation and copy number variation patterns of the risk score. (a, b) An analysis of the 20 most mutated genes of either
subgroup ((a), lower risk score; (b), higher risk score) was performed. Columns are sorted by samples with increasing risk score. (a) The sum
of mutations in each of the sample categories is indicated by the legend; (b) the sum of the mutations in each gene is indicated by the legend.
(c, d) The overall recurrent copy number variation (CNV) profile in order of increasing risk score ((c), lower risk score; (d), higher risk
score).

Aneuploidy, also known as somatic copy number al- results of these analyses indicated that a lower risk score
terations, is widespread in human cancers and has been was correlated with immune/inflammatory responses,
proposed to drive tumorigenesis. Aneuploidy was pre- and a higher risk score was associated with cell-cycle-
viously demonstrated to correlate with tumor cell pro- related processes. These results could partially explain
liferation and reduced immune processes [33]. In our that patients possessing a higher risk score demonstrated
study, overall copy number variant counts in high or low a worse prognosis, and patients with more active im-
risk score cases were not significantly different. This mune/inflammatory responses exhibited a better out-
suggested that our signature could infer immune/in- come. The high correlation between the risk score and the
flammatory responses irrespective of aneuploidy differ- tumor purity also suggested the presence of more immune
ence; however, some key genes did exhibit different cell infiltration in lower risk score cases. We also per-
variation patterns. The deletion of CDKN2A, a tumor formed ssGSEA analysis to evaluate the relationship
suppressor gene that functions in G1 cell cycle control, was between our risk score and the tumor purity as defined by
associated with poor prognosis and low survival rate in expression data. ImmuneScore, StromalScore, ESTI-
OC-SCC [34], and EGFR amplification was shown to be MATE score, and tumor purity were all significantly
associated with advanced clinical stage in OC-SCC pa- correlated with the risk score, and of these scores, the
tients [35]. The result of the aneuploidy difference re- ImmuneScore exhibited the highest correlation. Of note,
garding CDKN2A and EGFR was in accordance with the different from other cancer types such as glioma [36] and
worse prognosis higher risk score represented. colon cancer [37], the purity-related scores or purity
We performed GO and GSEA analyses to further alone were unable to distinguish among different prog-
validate the functional annotation of our signature. The nostic patient groups [38].
Journal of Oncology 7

Percent of enriched gene set


0 20 40 60 80 100 120

Immune response
Inflammatory response
Defense response
Regulation of lymphocyte activation
Positive regulation of immune system process
Leukocyte activation
Response to wounding
Regulation of cell activation
Positive regulation of cell activation
Cell activation
Regulation of leukocyte activation
Positive regulation of leukocyte activation
Chemotaxis
Taxis
Lymphocyte activation
Regulation of T-cell activation
Locomotory behavior
Behavior
Positive regulation of lymphocyte activation
T-cell activation
Antigen processing and presentation of peptide
or polysaccharide antigen via MHC class II
Positive regulation of response to stumulus
Cellular defense response
Cellular metal ion homeostasis
Cellular calcium ion homeostasis

0 20 40 60
–log10 (FDR)

(a)
Percent of enriched gene set
0 10 20 30 40

RNA processing translation


G−protein coupled receptor protein signaling pathway
Cell cycle
Cell cycle process
M phase of mitotic cell cycle
Mitosis
Nuclear division
DNA metabolic process
Mitotic cell cycle
Cell surface receptor linked signal transduction
RNA splicing
Nuclear mRNA splicing via spliceosome
RNA splicing via transesterfication
Reactions with bulged adenosine as nucleophile
RNA splicing via transesterification reactions
mRNA metabolic process
M phase
Ribonucleoprotein complex biogenesis
mRNA processing
Cell cycle phase
Organelle fision
DNA repair
Neurological system process
Cell division
ncRNA processing

0 5 10 15 20
–log10 (FDR)

(b)
Enrichment plot: Enrichment plot: Enrichment plot:
HALLMARK_INFLAMMATORY_RESPONSE KEGG_T_CELL_RECEPROR_SIGNALING_PATHWAY GO_LYMPHOCYTE_COSTIMULATION
0.0 0.0 0.0
Ranked list metric (Signal2Noise() Enrichment score (ES)

Ranked list metric (Signal2Noise() Enrichment score (ES)

Ranked list metric (Signal2Noise() Enrichment score (ES)

NES = –1.743 NES = –1.879 NES = –2.024


–0.1 –0.1 –0.1
FD 0.001 FDR < 0.001 FDR < 0.001
–0.2 –0.2 –0.2
–0.3 –0.3 –0.3
–0.4 –0.4 –0.4
–0.5 –0.5 –0.5
–0.6 –0.6 –0.6
–0.7 –0.7 –0.7

0.25 “1” (positively correlated) 0.25 “1” (positively correlated) 0.25 “1” (positively correlated)
0.00 0.00 0.00
–0.25 Zero cross at 9620 –0.25 Zero cross at 9620 –0.25 Zero cross at 9620
–0.50 –0.50 –0.50
–0.75 –0.75 –0.75
“0” (negatively correlated) “0” (negatively correlated) “0” (negatively correlated)
0 2.5000 5.0000 7.5000 10.000 12.500 15.000 17.500 20.000 0 2.5000 5.0000 7.5000 10.000 12.500 15.000 17.500 20.000 0 2.5000 5.0000 7.5000 10.000 12.500 15.000 17.500 20.000
Rank in ordered dataset Rank in ordered dataset Rank in ordered dataset

Enrichment profile Enrichment profile Enrichment profile


Hits Hits Hits
Ranking metric scores Ranking metric scores Ranking metric scores

(c)
Figure 4: Continued.
8 Journal of Oncology

Enrichment plot: Enrichment plot: Enrichment plot:


HALLMARK_MYC_TARGETS_V2 KEGG_DNA_REPLICATION GO_GENTROMERE_COMPLEX_ASSEMBLY
0.8
Enrichment score (ES)

Enrichment score (ES)

Enrichment score (ES)


0.5 NES = 2.338 0.6 NES = 2.445 0.7 NES = 3.110
FDR < 0.001 0.5 FDR < 0.001 0.6 FDR < 0.001
0.4
0.4 0.5
0.3 0.3 0.4
0.2 0.3
0.2
0.2
0.1
0.1 0.1
0.0 0.0
0.0
Ranked list metric (Signal2Noise()

Ranked list metric (Signal2Noise()

Ranked list metric (Signal2Noise()


0.25 “1” (positively correlated) 0.25 “1” (positively correlated) 0.25 “1” (positively correlated)
0.00 0.00 0.00
–0.25 Zero cross at 9620 –0.25 Zero cross at 9620 –0.25 Zero cross at 9620
–0.50 –0.50 –0.50
–0.75 “0” (negatively correlated) –0.75 “0” (negatively correlated) –0.75 “0” (negatively correlated)
0 2.5000 5.0000 7.5000 10.000 12.500 15.000 17.500 20.000 0 2.5000 5.0000 7.5000 10.000 12.500 15.000 17.500 20.000 0 2.5000 5.0000 7.5000 10.000 12.500 15.000 17.500 20.000
Rank in ordered dataset Rank in ordered dataset Rank in ordered dataset

Enrichment profile Enrichment profile Enrichment profile


Hits Hits Hits
Ranking metric scores Ranking metric scores Ranking metric scores

(d)

Figure 4: Biological annotation of the risk score. (a) GO results based on 1355 negatively correlated (R < − 0.4) genes. (b) GO results based
on 1632 positively correlated (R > 0.2) genes. (c) GSEA results of the lower risk score. (d) GSEA results of the higher risk score.

6000 <2.2e – 16 0.078 0.014 0.016 ANNOVA 2.4e – 11 ANOVA 1.6e – 05


6000
ImmuneScore

4000
4000

2000 2000

R = –0.707 0
0 P < 0.0001

–50 –40 –30 –20 –10


Low risk score

High risk score

Low risk score


Age < 60

Age ≥ 60

Female

Basal

Classical
Male

CpG island
Nesenchymal

Atypical
Stage I/II

Normal-like
Stage III/IV

Hypermethylated
Hypomethylated
Risk score

(a)
<2.2e – 16 0.63 0.59 0.46 ANNOVA 2.2e – 16 ANOVA 0.0017
4000 5000
StromalScore

2000 2500

0 0
R = –0.662
P < 0.0001
–2000
–50 –40 –30 –20 –10
Low risk score

High risk score

Age < 60

Age ≥ 60

Basal

Classical

Low risk score


Female

Male

CpG island
Nesenchymal

Atypical
Stage I/II

Normal-like
Stage III/IV

Hypermethylated
Hypomethylated

Risk score

(b)
Figure 5: Continued.
Journal of Oncology 9

1.00 <2.2e – 16 0.22 0.11 0.093 ANNOVA 2.2e – 16 ANOVA 2.6e – 04


0.75
0.75
Purity

0.50
0.50

0.25 0.25
R = 0.750
P < 0.0001
0.00 0.00
–50 –40 –30 –20 –10

Low risk score

High risk score

Age < 60

Age ≥ 60

Female

Classical

Low risk score


Male

CpG island
Nesenchymal

Atypical

Basal
Stage I/II

Normal-like
Stage III/IV

Hypermethylated
Hypomethylated
Risk score

(c)

Figure 5: Association between the risk score and tumor purity. (a) Correlation between the ImmnueScore and the risk score and the
distribution of the ImmnueScore among subgroups of OC-SCC within the training cohort. (b) Correlation between the StromalScore and
the risk score and the distribution of the StromalScore among subgroups of OC-SCC within the training cohort. (c) Correlation between
tumor purity and the risk score and the distribution of tumor purity among subgroups of OC-SCC in the training cohort.

5. Conclusion Supplementary Materials


Taken together, we identified and validated an eighteen- Figure S1: association between the risk score and the im-
gene-based immune/inflammatory signature that mune genes and association among genes in the signature.
exhibited independent prognostic significance for pa- Figure S2: distribution and prognostic value of the risk score
tients with OC-SCC and reflected the overall intensity of across different subgroups of the training cohort. Figure S3:
immune/inflammatory responses within the tumor mi- distribution and prognostic value of the risk score across
croenvironment. Our study offers new insights regarding different subgroups of the validation cohort. Figure S4:
the OC-SCC immune microenvironment and immune- association between the risk score and tumor purity in the
related therapy for this disease. Evaluating this signature validation cohort. Table S1: demographic characteristics in
may help to elucidate the complex role of tumor im- the training cohort and validation cohort. Table S2: risk
mune/inflammatory responses in this disease and will score formula. (Supplementary Materials)
provide new insight into clinical management and drug
design.
References
Data Availability [1] A. C. Chi, T. A. Day, and B. W. Neville, “Oral cavity and
oropharyngeal squamous cell carcinoma-an update,” CA: A
The preprocessed normalized expression data, CNV data, Cancer Journal for Clinicians, vol. 65, no. 5, pp. 401–421, 2015.
somatic mutation data, and clinical data of 314 OC-SCC [2] S. Gandini, E. Botteri, S. Iodice et al., “Tobacco smoking and
cancer: a meta-analysis,” International Journal of Cancer,
(training cohort) samples belong to the TCGA dataset, re-
vol. 122, no. 1, pp. 155–164, 2008.
trieved from the website http://cancergenome.nih.gov/. The
[3] R. A. Beynon, S. Lang, S. Schimansky et al., “Tobacco smoking
normalized expression data and clinical data of the and alcohol drinking at diagnosis of head and neck cancer and
GSE41613 dataset that comprised 97 OC-SCC samples were all-cause mortality: results from head and neck 5000, a
retrieved from the Gene Expression Omnibus website http:// prospective observational cohort of people with head and
www.ncbi.nlm.nih.gov/geo/query/acc.cgi?accGSE41613. neck cancer,” International Journal of Cancer, vol. 143, 2018.
[4] N. E Allen, J. Bénichou, K. Bloomfield et al., “Alcohol con-
Conflicts of Interest sumption and ethyl carbamate,” IARC Monographs on the
Evaluation of Carcinogenic Risks to Humans, vol. 96, pp. 3–
The authors declare that they have no conflicts of interest. 1383, 2010.
[5] L. Radoı̈, S. Paget-Bailly, D. Cyr et al., “Tobacco smoking,
alcohol drinking and risk of oral cavity cancer by subsite,”
Acknowledgments European Journal of Cancer Prevention, vol. 22, no. 3,
pp. 268–276, 2013.
The authors would like to thank Dr. Zhen-Hang Li for his [6] H. Li, S. J. Torabi, W. G. Yarbrough, S. Mehra, H. A. Osborn,
guidance in data analysis. This research was funded by the and B. Judson, “Association of human papillomavirus status
Tianjin Stomatological Hospital Key Project. This work was at head and neck carcinoma subsites with overall survival,”
supported by the Tianjin Key Discipline Foundation of JAMA Otolaryngology-Head & Neck Surgery, vol. 144, no. 6,
Clinical Medicine (HWZX002). pp. 519–525, 2018.
10 Journal of Oncology

[7] H. Mirghani, F. Amen, F. Moreau, and J. Lacau St Guily, “Do by the sequencing quality control consortium,” Nature Bio-
high-risk human papillomaviruses cause oral cavity squamous technology, vol. 32, no. 9, pp. 903–914, 2014.
cell carcinoma?,” Oral Oncology, vol. 51, no. 3, pp. 229–236, [24] A. Mayakonda and H. P. Koeffler, “Maftools: efficient analysis,
2015. visualization and summarization of MAF files from large-
[8] G. D’Souza, T. S. McNeel, and C. Fakhry, “Understanding scale cohort based cancer studies,” Genome Research, vol. 28,
personal risk of oropharyngeal cancer: risk-groups for on- 2016.
cogenic oral HPV infection and oropharyngeal cancer,” [25] A. Colaprico, T. C. Silva, C. Olsen et al., “TCGAbiolinks: an R/
Annals of Oncology: Official Journal of the European Society for Bioconductor package for integrative analysis of TCGA data,”
Medical Oncology, vol. 28, pp. 3065–3069, 2017. Nucleic Acids Research, vol. 44, no. 8, p. e71, 2016.
[9] C. R. Leemans, P. J. F. Snijders, and R. H. Brakenhoff, “The [26] K. Yoshihara, M. Shahmoradgoli, E. Martinez et al., “Inferring
molecular landscape of head and neck cancer,” Nature Re- tumour purity and stromal and immune cell admixture from
views Cancer, vol. 18, no. 5, pp. 269–282, 2018. expression data,” Nature Communications, vol. 4, p. 2612,
[10] E. Dediol, I. Sabol, M. Virag, M. Grce, D. Muller, and 2013.
S. Manojlović, “HPV prevalence and p16INKa overexpression [27] S. Choi and J. N. Myers, “Molecular pathogenesis of oral
in non-smoking non-drinking oral cavity cancer patients,” squamous cell carcinoma: implications for therapy,” Journal
Oral Diseases, vol. 22, no. 6, pp. 517–522, 2016. of Dental Research, vol. 87, no. 1, pp. 14–32, 2008.
[11] K. R. Sterba, E. Garrett-Mayer, M. J. Carpenter et al., [28] Y. Nishikawa, T. Miyazaki, K. Nakashiro et al., “Human FAT1
cadherin controls cell migration and invasion of oral squa-
“Smoking status and symptom burden in surgical head and
mous cell carcinoma through the localization of beta-cat-
neck cancer patients,” The Laryngoscope, vol. 127, no. 1,
enin,” Oncology Reports, vol. 26, pp. 587–592, 2011.
pp. 127–133, 2017.
[29] R. Li, R. Zhang, W. He, Y. Qiao, and W. Li, “The use of venous
[12] M. A. Saad, S. Z. Kuo, E. Rahimy et al., “Alcohol-dysregulated
coupler device in free tissue transfers for oral and maxillo-
miR-30a and miR-934 in head and neck squamous cell car-
facial reconstruction,” Journal of Oral and Maxillofacial
cinoma,” Molecular Cancer, vol. 14, p. 181, 2015. Surgery, vol. 73, no. 11, pp. 2225–2231, 2015.
[13] T. Barr, C. Helms, K. Grant, and I. Messaoudi, “Opposing [30] A. Chaturvedi, N. Husain, S. Misra et al., “Validation of the
effects of alcohol on the immune system,” Progress in Neuro- Brandwein Gensler risk model in patients of oral cavity
Psychopharmacology and Biological Psychiatry, vol. 65, squamous cell carcinoma in North India,” in Head and Neck
pp. 242–251, 2016. Pathology, Springer, New York, NY, USA, 2019.
[14] P. E. Molina, K. I. Happel, P. Zhang, J. K. Kolls, and S. Nelson, [31] Q. F. Jin, Q. G. Fang, J. X. Qi, and P. Li, “Impact of BMI on
“Focus on: alcohol and the immune system,” Alcohol Research complications and satisfaction in patients with papillary
& Health: The Journal of the National Institute on Alcohol thyroid cancer and lateral neck metastasis. Cancer control,”
Abuse and Alcoholism, vol. 33, pp. 97–108, 2010. Journal of the Moffitt Cancer Center, vol. 26,
[15] M. R. Stämpfli and G. P. Anderson, “How cigarette smoke p. 1073274819853831, 2019.
skews immune responses to promote infection, lung disease [32] D. M. Vossen, C. V. M. Verhagen, M. Verheij,
and cancer,” Nature Reviews Immunology, vol. 9, no. 5, L. F. A. Wessels, C. Vens, and M. W. M. van den Brekel,
pp. 377–384, 2009. “Comparative genomic analysis of oral versus laryngeal and
[16] R. L. Ferris, “Immunology and immunotherapy of head and pharyngeal cancer,” Oral Oncology, vol. 81, pp. 35–44, 2018.
neck cancer,” Journal of Clinical Oncology, vol. 33, no. 29, [33] S. Park, F. Supek, and B. Lehner, “Systematic discovery of
pp. 3293–3304, 2015. germline cancer predisposition genes through the identifi-
[17] C. Badoual, S. Hans, N. Merillon et al., “PD-1-expressing cation of somatic second hits,” Nature Communications,
tumor-infiltrating T cells are a favorable prognostic biomarker vol. 9, p. 2601, 2018.
in HPV-associated head and neck cancer,” Cancer Research, [34] S. S. Padhi, S. Roy, M. Kar et al., “Role of CDKN2A/p16
vol. 73, no. 1, pp. 128–138, 2013. expression in the prognostication of oral squamous cell
[18] A. E. Grulich, M. T. van Leeuwen, M. O. Falster, and carcinoma,” Oral Oncology, vol. 73, pp. 27–35, 2017.
C. M. Vajdic, “Incidence of cancers in people with HIV/AIDS [35] V. Costa, L. P. Kowalski, C. M. Coutinho-Camillo et al.,
compared with immunosuppressed transplant recipients: a “EGFR amplification and expression in oral squamous cell
meta-analysis,” The Lancet, vol. 370, no. 9581, pp. 59–67, 2007. carcinoma in young adults,” International Journal of Oral and
[19] D. S. Michaud, S. M. Langevin, M. Eliot et al., “Allergies and Maxillofacial Surgery, vol. 47, no. 7, pp. 817–823, 2018.
risk of head and neck cancer,” Cancer Causes & Control, [36] C. Zhang, W. Cheng, X. Ren et al., “Tumor purity as an
vol. 23, no. 8, pp. 1317–1322, 2012. underlying key factor in glioma,” Clinical Cancer Research,
[20] N. C. G. Atlas, “Comprehensive genomic characterization of vol. 23, no. 20, pp. 6279–6291, 2017.
head and neck squamous cell carcinomas,” Nature, vol. 517, [37] Y. Mao, Q. Feng, P. Zheng et al., “Low tumor purity is as-
pp. 576–582, 2015. sociated with poor prognosis, heavy mutation burden, and
[21] P. Lohavanichbutr, E. Mendez, F. C. Holsinger et al., “A 13- intense immune phenotype in colon cancer,” Cancer Man-
gene signature prognostic of HPV-negative OSCC: discovery agement and Research, vol. 10, pp. 3569–3577, 2018.
and external validation,” Clinical Cancer Research, vol. 19, [38] S. Bai, P. Zhang, J.-C. Zhang et al., “A gene signature asso-
no. 5, pp. 1197–1203, 2013. ciated with prognosis and immune processes in head and neck
[22] A. Subramanian, P. Tamayo, V. K. Mootha et al., “Gene set squamous cell carcinoma,” Head & Neck, vol. 41, no. 8,
pp. 2581–2590, 2019.
enrichment analysis: a knowledge-based approach for inter-
preting genome-wide expression profiles,” Proceedings of the
National Academy of Sciences, vol. 102, no. 43, pp. 15545–
15550, 2005.
[23] Z Su, P. Łabaj, S. Li et al., “A comprehensive assessment of
RNA-seq accuracy, reproducibility and information content
MEDIATORS of

INFLAMMATION

The Scientific Gastroenterology Journal of


World Journal
Hindawi Publishing Corporation
Research and Practice
Hindawi
Hindawi
Diabetes Research
Hindawi
Disease Markers
Hindawi
www.hindawi.com Volume 2018
http://www.hindawi.com
www.hindawi.com Volume 2018
2013 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018

Journal of International Journal of


Immunology Research
Hindawi
Endocrinology
Hindawi
www.hindawi.com Volume 2018 www.hindawi.com Volume 2018

Submit your manuscripts at


www.hindawi.com

BioMed
PPAR Research
Hindawi
Research International
Hindawi
www.hindawi.com Volume 2018 www.hindawi.com Volume 2018

Journal of
Obesity

Evidence-Based
Journal of Stem Cells Complementary and Journal of
Ophthalmology
Hindawi
International
Hindawi
Alternative Medicine
Hindawi Hindawi
Oncology
Hindawi
www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2013

Parkinson’s
Disease

Computational and
Mathematical Methods
in Medicine
Behavioural
Neurology
AIDS
Research and Treatment
Oxidative Medicine and
Cellular Longevity
Hindawi Hindawi Hindawi Hindawi Hindawi
www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018

You might also like