Skip to main content
Molecular Oncology logoLink to Molecular Oncology
. 2018 Oct 26;12(12):2085–2101. doi: 10.1002/1878-0261.12388

A prognostic mRNA expression signature of four 16q24.3 genes in radio(chemo)therapy‐treated head and neck squamous cell carcinoma (HNSCC)

Ludmila Wintergerst 1,2, Martin Selmansberger 1, Cornelius Maihoefer 2,3, Lars Schüttrumpf 2,3, Axel Walch 4, Christina Wilke 1, Adriana Pitea 1,5, Christine Woischke 6, Philipp Baumeister 2,7, Thomas Kirchner 6, Claus Belka 2,3,8, Ute Ganswindt 2,3,9, Horst Zitzelsberger 1,2,3, Kristian Unger 1,2,3, Julia Hess 1,2,3,
PMCID: PMC6275282  PMID: 30259648

Abstract

Previously, we have shown that copy number gain of the chromosomal band 16q24.3 is associated with impaired clinical outcome of radiotherapy‐treated head and neck squamous cell carcinoma (HNSCC) patients. We set out to identify a prognostic mRNA signature from genes located on 16q24.3 in radio(chemo)therapy‐treated HNSCC patients of the TCGA (The Cancer Genome Atlas, n = 99) cohort. We applied stepwise forward selection using expression data of 41 16q24.3 genes. The resulting optimal Cox‐proportional hazards regression model included the genes APRT, CENPBD1, CHMP1A, and GALNS. Afterward, the prognostic value of the classifier was confirmed in an independent cohort of HNSCC patients treated by adjuvant radio(chemo)therapy (LMU‐KKG cohort). The signature significantly differentiated high‐ and low‐risk patients with regard to overall survival (HR = 2.01, 95% CI 1.10–3.70; P = 0.02125), recurrence‐free survival (HR = 1.84, 95% CI 1.01–3.34; P = 0.04206), and locoregional recurrence‐free survival (HR = 1.87, 95% CI 1.03–3.40; P = 0.03641). The functional impact of the four signature genes was investigated after reconstruction of a gene association network from transcriptome data of the TCGA HNSCC cohort using a partial correlation approach. Subsequent pathway enrichment analysis of the network neighborhood (first and second) of the signature genes suggests involvement of HNSCC‐associated signaling pathways such as apoptosis, cell cycle, cell adhesion, EGFR, JAK‐STAT, and mTOR. Furthermore, a detailed analysis of the first neighborhood revealed a cluster of co‐expressed genes located on chromosome 16q, substantiating the impact of 16q24.3 alterations in poor clinical outcome of HNSCC. The reported gene expression signature represents a prognostic marker in HNSCC patients following postoperative radio(chemo)therapy.

Keywords: classifier, gene association network, head and neck squamous cell carcinoma, prognosis, radiotherapy, signature


Abbreviations

API

Application Programming Interface

APRT

adenine phosphoribosyltransferase

CENPBD1

CENPB DNA‐binding domain containing 1

CHMP1A

charged multivesicular body protein 1A

CI

confidence interval

FDR

false discovery rate

FFPE

formalin‐fixed paraffin‐embedded

GALNS

galactosamine (N‐acetyl)‐6‐sulfatase

GAN

gene association network

GSEA

gene set enrichment analysis

HE

hemotoxylin and eosin

HNSCC

head and neck squamous cell carcinoma

HPV

human papilloma virus

HR

hazard ratio

IHC

immunohistochemistry

NE

not estimable

OS

overall survival

PGK1

phosphoglycerate kinase 1

q‐PCR

quantitative real‐time polymerase chain reaction

qRT‐PCR

quantitative real‐time reverse transcription polymerase chain reaction

SNP

single nucleotide polymorphisms

TPM

Transcripts Per Million

1. Introduction

Head and neck squamous cell carcinoma (HNSCC) comprises a group of heterogeneous tumors from different anatomical sites with tobacco smoking and alcohol abuse as the major risk factors (Marcu and Yeoh, 2009). Infection with high‐risk human papillomavirus (HPV) is another emerging risk factor. HPV‐related tumors are characterized by a distinct molecular pathogenesis with a considerably favorable prognosis (Leemans et al., 2011; O'Sullivan et al., 2013). Despite advances in treatment modalities, the 5‐year survival rate for advanced HNSCC still needs improvement. The identification of patients with therapy‐resistant tumors using prognostic markers would allow personalized treatment approaches (Mirghani et al., 2015; O'Sullivan et al., 2013); however, such markers are not yet established.

So far, research groups have mainly focused on the identification of single molecular markers to be used for diagnosis and treatment selection in HNSCC (Rocco et al., 2006). Due to the complexity and heterogeneity of tumors, it is likely that instead of a single gene, the prognostic value of a gene signature with regard to patient outcome would be more powerful (Ginos et al., 2004; Lohavanichbutr et al., 2013; Pavon et al., 2012). Up to now, mRNA expression signatures associated with metastasis (Lian et al., 2013; Roepman et al., 2006), hypoxia (Eustace et al., 2013; Toustrup et al., 2012), HPV status (Cancer Genome Atlas, 2015; Slebos et al., 2006), and immune response (Chung et al., 2004; Wood et al., 2016) have been reported in HNSCC. Although, for example, the immune response signature of Chung et al. and the hypoxia signatures developed by Eustace et al. and Toustrup et al. could be confirmed in subsequent publications (Keck et al., 2015; Tawk et al., 2016), many molecular signatures, some of them very complex, fail independent validation and therefore to change practice in a clinical setting. This might be explained by methodological aspects such as the selection and number of genes examined, differences in the analysis platforms used, restrictions due to small sample sizes, lack of independent validation, but also by demographic differences in the patient groups examined or the unavailability of detailed clinical information.

Previously, we demonstrated an association of gains of chromosomal band 16q24.3 with locoregional progression‐free survival of radiotherapy‐treated HNSCC patients (Bauer et al., 2008) and validated this marker in the subgroup of adjuvant radio(chemo)therapy‐treated patients of the TCGA HNSCC cohort (Hess et al., 2017). Since copy number alterations might lead to aberrant mRNA expression of genes (Gollin, 2014), our objective was to develop a prognostic mRNA signature from genes located on this chromosomal band using a stepwise forward selection approach. The prognostic value of the gene classifier was analyzed in an independent HNSCC cohort. In addition, the functional role of the signature genes was investigated.

2. Methods

2.1. Patients

Our study was conducted in compliance with the Reporting recommendations for tumor MARKer prognostic studies (REMARK) (McShane et al., 2005) and fulfills the requirements set out by Simon et al. (2009), which are summarized in Table S1 (Simon et al., 2009).

We analyzed two independent cohorts of HNSCC patients who had undergone surgical resection followed by adjuvant radio(chemo)therapy: the TCGA and the LMU‐KKG cohort (Ludwig‐Maximilians‐University of Munich, Clinical Cooperation Group ‘Personalized Radiotherapy in Head and Neck Cancer’) (Maihoefer et al., 2018).

For the TCGA HNSCC cohort, mRNA expression (RNA Seq V2 RSEM)‐level z‐scores of genes located on chromosomal band 16q24.3 (generated by the TCGA Research Network http://cancergenome.nih.gov) were downloaded from cBioPortal (2015/08/12) (Cancer Genome Atlas, 2015; Cerami et al., 2012; Gao et al., 2013). Inclusion criteria for patients of the TCGA cohort were as follows: (a) Treatment with radio(chemo)therapy; (b) no therapy in the frame of neoadjuvant, recurrent, or palliative treatment; (c) availability of HPV status; (d) availability of mRNA expression and genomic copy number data of genes located on chromosomal band 16q24.3. The resulting patient subset of 99 radio(chemo)therapy‐treated HNSCC patients (Table S2) was randomly split into a training (n = 40) and a validation set (n = 59) while ensuring equal percentage distribution of HPV‐positive cases. Median follow‐up times of the training and validation set were 656 and 643 days, respectively.

The retrospective LMU‐KKG cohort served as an independent validation cohort. This study on clinical and biological data was approved by the local ethics committee in Munich (EA 448‐13 and 17‐116) and carried out in accordance with the Declaration of Helsinki. The experiments were undertaken with the understanding and written consent of each subject. The LMU‐KKG cohort included all patients with HNSCC of the hypopharynx, larynx, oropharynx, or oral cavity treated at the Department of Radiation Oncology, LMU, Germany, between 2008 and 2013 (Maihoefer et al., 2018). All patients received adjuvant radiotherapy as a curative approach after surgical resection. The median overall treatment time was 45 days (interquartile range 43–47 days) with five fractions per week. A median radiation dose of 64 Gy (2 Gy/fraction) was applied to the former tumor bed or regions of ECE. Elective lymph node regions have been covered according to tumor stage and localization with a median dose of 50 Gy (2 Gy/fraction) and 56 Gy (2 Gy/fraction) were applied to involved lymph node regions. In the case of close (R0, but less than 5 mm) or positive microscopic resection margins and/or ECE, patients received concurrent chemotherapy; 47.2% of the patients received CDDP/5‐fluorouracil (5‐FU) (CDDP: 20 mg·m−2 days 1–5/29–33; 5‐FU: 600 mg·m−2 days 1–5/29–33). In some cases, Mitomycin C (MMC) (10.2%) or 5‐FU/MMC (5.6%) was used instead of platinum‐based chemotherapy. End of follow‐up period was on 14 May 2016, and the median OS time was 1878 days. The clinical and pathological data for both cohorts are presented in Table 1. A comparison of demographic parameters between the TCGA and the LMU‐KKG cohort is given in Table S3.

Table 1.

Demographic and treatment characteristics of the total adjuvant radio(chemo)therapy‐treated TCGA HNSCC cohort, of the TCGA HNSCC training and validation set and the adjuvant radio(chemo)therapy‐treated LMU‐KKG HNSCC patient cohort and stratified according to the four‐gene signature. % represents percentage of all patients

TCGA cohort all (n = 99) TCGA training set (n = 40) TCGA validation set (n = 59) LMU‐KKG cohort (n = 108)
No. of all patients No. of all patients Low‐risk (n = 24) High‐risk (n = 16) P‐valuea No. of all patients Low‐risk (n = 30) High‐risk (n = 29) P‐valuea No. of all patients Low‐risk (n = 53) High‐risk (n = 55) P‐valuea
Sex, no. (%)
Male 79 (80) 35 (88) 22 (92) 13 (81) 0.37 44 (75) 24 (80) 20 (69) 0.38 74 (69) 37 (70) 37 (67) 0.84
Female 20 (20) 5 (13) 2 (8) 3 (19) 15 (25) 6 (20) 9 (31) 34 (31) 16 (30) 18 (33)
Age (years), no. (%)
< 60 56 (57) 23 (58) 12 (50) 11 (69) 0.33 33 (56) 16 (53) 17 (59) 0.79 44 (41) 25 (47) 19 (35) 0.24
≥ 60 43 (43) 17 (43) 12 (50) 5 (31) 26 (44) 14 (47) 12 (41) 64 (59) 28 (53) 36 (65)
Tumor localization, no. (%)
Oral cavity 49 (49) 17 (43) 8 (33) 9 (56) 0.067 32 (54) 14 (47) 18 (62) 0.51 27 (25) 14 (26) 13 (24) 0.91
Larynx 28 (28) 14 (35) 8 (33) 6 (38) 14 (24) 8 (27) 6 (21) 12 (11) 6 (11) 6 (11)
Oropharynx 20 (20) 7 (18) 7 (29) 0 (0) 13 (22) 8 (27) 5 (17) 52 (48) 26 (49) 26 (47)
Hypopharynx 2 (2) 2 (5) 1 (4) 1 (6) 0 (0) 0 (0) 0 (0) 17 (16) 7 (13) 10 (18)
Alcohol consumption, no. (%)
Yes 84 (85) 33 (83) 19 (79) 14 (88) 0.38 51 (86) 25 (83) 26 (90) 1 53 (49) 27 (51) 26 (47) 1
No 13 (13) 6 (15) 5 (21) 1 (6) 7 (12) 4 (13) 3 (10) 32 (30) 16 (30) 16 (29)
Missing information 2 (2) 1 (3) 0 (0) 1 (6) 1 (2) 1 (3) 0 (0) 23 (21) 10 (19) 13 (24)
Smoking status, no. (%)
Smoker 80 (81) 30 (75) 16 (67) 14 (88) 0.26 50 (85) 26 (87) 24 (83) 0.73 71 (66) 29 (55) 42 (76) 1
Never smoker 19 (19) 10 (25) 8 (33) 2 (13) 9 (15) 4 (13) 5 (17) 9 (8) 4 (8) 5 (9)
Missing information 0 (0) 0 (0) 0 (0) 0 (0) 0 (0) 0 (0) 0 (0) 28 (26) 20 (38) 8 (15)
UICC TNM stage, no. (%)
I 2 (2) 0 (0) 0 (0) 0 (0) 0.84 2 (3) 2 (7) 0 (0) 0.089 1 (1) 0 (0) 1 (2) 0.92
II 4 (4) 3 (8) 2 (8) 1 (6) 1 (2) 1 (3) 0 (0) 10 (9) 5 (9) 5 (9)
III 7 (7) 1 (3) 1 (4) 0 (0) 6 (10) 1 (3) 5 (17) 33 (31) 18 (34) 15 (27)
IVa 57 (58) 23 (58) 14 (58) 9 (56) 34 (58) 18 (60) 16 (55) 61 (56) 29 (55) 32 (58)
IVb 1 (1) 1 (3) 0 (0) 1 (6) 0 (0) 0 (0) 0 (0) 3 (3) 1 (2) 2 (4)
Missing information 28 (28) 12 (30) 7 (29) 5 (31) 16 (27) 8 (27) 8 (28) 0 (0) 0 (0) 0 (0)
pT stage, no. (%)
T1 5 (5) 1 (3) 1 (4) 0 (0) 1 4 (7) 2 (7) 2 (7) 0.59 19 (18) 10 (19) 9 (16) 0.31
T2 18 (18) 9 (23) 6 (25) 3 (19) 9 (15) 4 (13) 5 (17) 45 (42) 18 (34) 27 (49)
T3 13 (13) 3 (8) 2 (8) 1 (6) 10 (17) 7 (23) 3 (10) 29 (27) 18 (34) 11 (20)
T4 37 (7) 17 (43) 10 (42) 7 (44) 20 (34) 9 (30) 11 (38) 15 (14) 7 (13) 8 (15)
Missing information 26 (26) 10 (25) 5 (21) 5 (31) 16 (27) 8 (27) 8 (28) 0 (0) 0 (0) 0 (0)
pN stage, no. (%)
N0 20 (20) 9 (23) 4 (17) 5 (31) 0.63 11 (19) 5 (17) 6 (21) 1 31 (29) 16 (30) 15 (27) 0.95
N1 11 (11) 3 (8) 2 (8) 1 (6) 8 (14) 4 (13) 4 (14) 26 (24) 7 (13) 13 (24)
N2 39 (39) 16 (40) 11 (46) 5 (31) 23 (39) 12 (40) 11 (38) 49 (45) 23 (43) 26 (47)
N3 0 (0) 0 (0) 0 (0) 0 (0) 0 (0) 0 (0) 0 (0) 2 (2) 1 (2) 1 (2)
Missing information 29 (29) 12 (30) 7 (29) 5 (31) 17 (29) 9 (30) 8 (28) 0 (0) 0 (0) 0 (0)
Grade, no. (%)
G1 (well differentiated) 5 (5) 0 (0) 0 (0) 0 (0) 0.74 5 (8) 3 (10) 2 (7) 0.64 3 (3) 3 (6) 0 (0) 0.11
G2 (moderate differentiated) 62 (63) 25 (63) 14 (58) 11 (69) 37 (63) 16 (53) 21 (72) 37 (34) 15 (28) 22 (40)
G3 (poorly differentiated) 29 (29) 15 (38) 10 (42) 5 (31) 14 (24) 8 (27) 6 (21) 68 (63) 35 (66) 33 (60)
Missing information 3 (3) 0 (0) 0 (0) 0 (0) 3 (5) 3 (10) 0 (0) 0 (0) 0 (0) 0 (0)
HPV status, no. (%)
Positive 21 (21) 9 (23) 9 (38) 0 (0) 0.006 12 (20) 11 (37) 1 (3) 0.00247 23 (21) 10 (19) 13 (24) 0.64
Negative 78 (79) 31 (78) 15 (62) 16 (100) 47 (80) 19 (63) 28 (97) 85 (79) 43 (81) 42 (76)
Simultaneous chemotherapy, no. (%)
Platinum‐based 55 (56) 22 (55) 15 (63) 7 (44) 0.29 33 (56) 18 (60) 15 (52) 0.15 54 (50) 28 (53) 26 (47) 0.71
Other 4 (4) 1 (3) 0 (0) 1 (6) 3 (5) 3 (10) 0 (0) 18 (17) 7 (13) 11 (20)
No 26 (26) 8 (20) 4 (17) 4 (25) 18 (31) 7 (23) 11 (38) 36 (33) 18 (34) 18 (33)
Missing information 14 (14) 9 (23) 5 (21) 4 (25) 5 (8) 2 (7) 3 (10) 0 (0) 0 (0) 0 (0)
a

Chi‐square test or Fisher's exact test.

Hemotoxylin and eosin (HE)‐stained tissue sections from available formalin‐fixed and paraffin‐embedded (FFPE) tissue blocks were histopathologically reviewed by a pathologist (A.W.), and the tumor containing area was defined. Samples with < 50% tumor cells were excluded from further analysis. Guided by the HE‐stained tissue slides, the annotated tumor area was micro‐dissected followed by simultaneous DNA and RNA extraction using the AllPrep DNA/RNA FFPE Kit (Qiagen, Hilden, Germany) according to the manufacturer's protocol. After DNA/RNA isolation and quality assessment, 108 HNSCC samples remained for further analysis.

2.2. Determination of the HPV status

HPV status of the patients was determined by p16INK4a immunohistochemistry in combination with HPV DNA detection. Immunohistochemical (IHC) p16INK4a staining, used as a surrogate marker for HPV‐infection, was performed using the CINtec TM Histology Kit (Roche mtm Laboratories AG, Heidelberg, Germany) on a Ventana Benchmark LT automated immunostainer (Ventana Medical Systems, Tucson, AZ, USA) according to the manufacturer's protocol. FFPE sections of embedded p16‐positive (UPCI SCC154) and p16‐negative HNSCC cell lines (Cal33) were included as positive and negative controls. Tumor specimens with strong and diffuse nuclear and cytoplasmic staining in more than 70% of tumor cells were considered as p16‐positive, whereas tissues with only faintly diffuse or absent staining were considered as p16‐negative (Ang et al., 2010). p16‐stained tissue sections were evaluated by two independent observers (L.W. and J.H.). Further, detection of mucosotropic HPV DNA was performed using quantitative real‐time PCR (q‐PCR) in combination with SYBR green chemistry (Clontech Laboratories, Inc., Mountain View, CA, USA). DNA samples (50 ng) were subjected to q‐PCR products (10 μL) on a ViiA 7 q‐PCR system (Thermo Fisher Scientific, Dreieich, Germany) using GP5+/6+ primers detecting the L1 gene (Eurofins MWG Operon, Ebersberg, Germany) (forward primer: 5′‐TTTGTTACTGTGGTAGATACTAC‐3′, reverse primer: 5′‐GAAAAATAAACTGTAAATCATATTC‐3′; amplicon size: 142 bp) (Hesselink et al., 2005). The β‐globin gene served as quality control (forward primer: 5′‐CAGGTACGGCTGTCATCACTTAGA‐3′, reverse primer: 5′‐CATGGTGTCTGTTTGAGGTTGCTA‐3′; amplicon size: 185 bp) (Metabion International AG, Planegg‐Martinsried, Germany) (Lindh et al., 2007). Two HPV‐positive (UPCI SCC2 and UPCI SCC154) and two HPV‐negative (Cal27 and Cal33) cell lines were included as controls. Reactions were carried out in triplicates along with negative controls. Samples with a detectable β‐globin PCR product (Ct‐value < 35) were considered as HPV‐negative if no HPV amplification product was detectable.

A tumor specimen was classified as HPV‐positive if it was positive for both p16INK4a IHC and HPV DNA status assessed by GP 5+/6+ q‐PCR (Smeets et al., 2007).

2.3. Development of a gene classifier

A schematic workflow for the development of the gene classifier and the reconstruction of the gene association network (GAN) with subsequent analyses is presented in Fig. S1.

A prognostic gene classifier regarding OS was built by applying a robust likelihood‐based survival modeling approach on mRNA gene expression data (z‐scores) of the TCGA training set using the R‐package rbsurv (Cho et al., 2009). A stepwise forward selection algorithm computed the partial likelihood of the Cox‐proportional hazards regression model for a sequential selection of mRNA (100 iterations, twofold cross‐validation), which allowed choosing the best performing model based on the Akaike information criterion, an estimator of the relative quality of a statistical model considering the goodness of fit penalized by model complexity, in terms of the best trade‐off between minimum complexity and best goodness of fit of the model.

Cox model coefficients [adenine phosphoribosyltransferase (APRT): 1.20988822, CENPBD1: 0.06012163, CHMP1A: 0.17153750, GALNS: 0.22431583] were multiplied with the corresponding mRNA expression values and summed up to an individual risk score for each patient. For the determination of the cutoff for the individual risk score stratifier, survival analyses were performed in the TCGA training set with cutoff values varying with increment of 0.1 starting from the minimal risk score. The cutoff, which resulted in the optimal split of patients in terms of log‐rank test P‐value (OS) in the training set, was used for assorting each patient individually into a low‐risk (< −0.2932616) and a high‐risk group (≥ −0.2932616). Both the model and the cutoff value derived from the TCGA training dataset were applied to the expression data of the TCGA validation set and the LMU‐KKG cohort. Expression data for the LMU‐KKG cohort were gained from quantitative real‐time RT‐PCR (qRT‐PCR) analysis.

2.4. Quantitative real‐time RT‐PCR (qRT‐PCR) analysis of the signature genes in the LMU‐KKG cohort

For the LMU‐KKG HNSCC cohort, mRNA expression of the signature genes was analyzed by qRT‐PCR. Reverse transcription was performed using the SuperScript® VILO™ cDNA Synthesis Kit (Life Technologies, Carlsbad, CA, USA) with 500 ng RNA input. qRT‐PCR products (10 μL) were carried out in triplicates according to manufacturer's protocol on an Applied Biosystems® ViiA™7 platform employing the Taqman® Assays (Life Technologies) APRT‐Hs00975725_m1, CHMP1A‐Hs00946132_g1, CENP BD1‐Hs00924894_s1, and GALNS‐Hs00975732_m1. β‐Actin (ACTB‐Hs01060665_g1) and phosphoglycerate kinase 1 (PGK1‐Hs99999906_m1) served as endogenous controls. Expression levels were determined using the ΔCt method followed by z‐score transformation.

2.5. Clinical endpoints LMU‐KKG cohort

Clinical endpoints included OS, recurrence‐free survival, and locoregional recurrence‐free survival. OS was calculated (in days) from the date of radiotherapy treatment start to the date of death from any cause. Recurrence‐free survival was defined as the time (days) from the start of radio(chemo)therapy treatment to the first observation of locoregional/distant recurrence or death due to any cause; locoregional recurrence‐free survival from the start of radio(chemo)therapy treatment to the date of local recurrence or death due to any cause. In the absence of an event, patients were censored at the date of the last follow‐up visit.

2.6. Genomic copy number data

File IDs of patients of the adjuvant radio(chemo)therapy‐treated TCGA HNSCC cohort were extracted using the GDC web API. Genomic copy number alterations SNP 6.0 raw data (.CEL files) were downloaded from the GDC data portal (https://portal.gdc.cancer.gov/). The CEL files were batch processed using functions of the rawcopy R‐package (http://rawcopy.org). The normal tissue data served as reference data to build normalized log2 intensity copy number ratios for each of the tumor samples. The log2‐ratios were segmented and the copy number status determined using functions of the R‐package CGHcall (van de Wiel et al., 2007). After determination of the DNA status, integration analysis of copy number data with the transcriptome data was carried out.

2.7. Statistical analysis

Comparisons of two Kaplan–Meier curves were performed using the log‐rank test of the R‐package survival; P‐values < 0.05 were considered statistically significant. Median estimates and hazard ratios (HR) with 95% confidence intervals (CI) were determined. P‐values were also calculated for the training set, but since no valid null‐hypothesis can be formulated in this case, it only reflects the meaningful split of risk groups. The association of clinical parameters with clinical endpoints was assessed using univariate Cox‐proportional hazards regression analysis. Parameters with P < 0.05 in univariate analysis were included into a multivariate Cox‐proportional hazards model.

Association of the copy number status of chromosomal band 16q24.3 with risk scores or mRNA expression levels of the signature genes was evaluated using unpaired two‐sided Mann–Whitney U‐test.

2.8. Gene association network (GAN) reconstruction

Raw RNA sequencing data on 98 HNSCC cases of the adjuvant radio(chemo)therapy‐treated TCGA HNSCC cohort were downloaded from the GDC data portal (https://portal.gdc.cancer.gov/). The RNA sequencing reads contained in the BAM files were realigned to the reference transcriptome (GRCH38/Ensembel) and quantified as Transcripts Per Million (TPM) using the tool Salmon (Patro et al., 2017). The identification of differentially expressed genes between the high‐risk and low‐risk group was carried out using the R‐package DEseq2, where genes with a cumulative (over samples) TPM ≤ 5 were excluded and genes with an adjusted P‐value < 0.1 were considered statistically significant (Love et al., 2014).

Differentially expressed genes were subsequently subjected to GAN reconstruction using the method implemented in the GeneNet R‐package, which is based on regularized dynamic partial correlation (Opgen‐Rhein and Strimmer, 2007). The edge probability cutoff of 0.99 was applied to obtain the resulting undirected GAN.

2.9. Gene set enrichment analysis

Gene sets from the Reactome database for gene set enrichment analysis (GSEA) were downloaded from the Broad Institute Molecular Signatures Database. GSEA was carried out in the preranked mode: All genes of the data set were ranked according to the log2 transformed fold changes (four‐gene signature high‐risk versus low‐risk group). GSEA tests for up‐ or down‐regulation of gene sets (pathways), while gene sets with false discovery rate (FDR) < 0.05 were considered statistically significant.

2.10. Pathway enrichment analysis

The Cytoscape Reactome Functional Interaction (FI) plugin (version 3.5.1) was used to perform a network clustering of the FI network consisting of genes from the GANs first and second neighborhood of the four signature genes (Shannon et al., 2003; Wu et al., 2010). A pathway enrichment analysis was conducted for modules including more than 100 genes. Pathways containing less than 200 but more than 20 genes were considered for pathway enrichment analysis. P‐values were determined by one‐sided Fisher's exact test. Benjamini–Hochberg adjusted P‐values (FDR) < 0.05 were considered statistically significant and ranked according to ascending FDRs.

2.11. Integration of copy number data with the transcriptome data

In order to assess whether mRNA expressions were explainable by genomic copy number alterations, an integration analysis was carried out on 98 HNSCC samples from the TCGA cohort. For this purpose, the Gene wise cisTest implemented in the Bioconductor sigaR R‐package was used (van Wieringen and van de Wiel, 2009; van Wieringen et al., 2012). Default values provided by the package were applied during the different analysis steps for all parameters, except nGenes = 500, which potentially improves the overall power of the FDR procedure included in the cisEffectTune function. Overall, this integrative analysis assesses the effect of the genomic copy number status of each gene on its mRNA expression level and provides a measure on how much of the mRNA expression is explained by its copy number status. The results are based on a permutation approach, while 10 000 permutations were used. The resulted P‐values were adjusted for multiple testing with the Benjamini–Hochberg method. Due to the nature of permutation tests, the results vary upon repetition of the test but stay stable with the respect to the drawn conclusions.

3. Results

The identified best performing prognostic model generated from mRNA expression data of 16q24.3 genes (n = 41) included the four genes APRT, CENPBD1, CHMP1A, and GALNS allowing to stratify HNSCC patients with regard to OS. A 5.76‐fold (95% CI 1.73–19.17) increased risk for death was observed for high‐risk patients (9/16, 56% patients with events) compared to the low‐risk group (7/24, 29% patients with events) of the TCGA training set (n = 40) (Fig. 1A). The robustness of the four‐gene classifier was confirmed in the TCGA validation set as high‐risk classified patients showed significantly reduced OS rates (10/29, 34% patients with events; hazard ratio (HR) 3.81, 95% CI 1.05–13.89; P = 0.02911; Fig. 1B) compared to low‐risk patients (3/30, 10% patients with events).

Figure 1.

Figure 1

Identification of a four‐gene classifier predicting OS in the subgroup of radio(chemo)therapy‐treated HNSCC patients of the TCGA cohort. (A) Kaplan–Meier curves for the endpoint OS for patients stratified according to the four‐gene classifier of the training (a) and validation (b) set within the radio(chemo)therapy‐treated TCGA HNSCC cohort. Survival curves are depicted for all cases (training set: n = 40, validation set: n = 59) and for HPV‐negative cases only (training set: n = 31, validation set: n = 47). P‐values, median OS times, and hazard ratios (HR) with 95% contingency intervals were obtained by Log‐rank test and are indicated. (B) Heatmap of mRNA expression levels (z‐scores) of the four signature genes (top panel) arranged according to risk scores (bottom panel) for the training (a) and validation (b) set within the TCGA HNSCC cohort. mRNA expression levels of all four signature genes were elevated in patients of the high‐risk group. The results are in whole based upon data generated by the TCGA Research Network: http://cancergenome.nih.gov (Cancer Genome Atlas, 2015; Cerami et al., 2012; Gao et al., 2013). NE, not estimable.

The mRNA expression levels of all signature genes were up‐regulated in high‐risk patients and correlated positively with the defined risk groups (Fig. 1A,B, lower panel).

The four‐gene classifier was independently validated in the LMU‐KKG cohort (n = 108) as high‐risk group patients showed significantly impaired OS rates (29/55, 45% patients with events; HR 2.01, 95% CI 1.10–3.70; P = 0.02125) compared to low‐risk patients (17/53, 32% patients with events) (Fig. 2A). Moreover, the risk groups significantly differed with regard to locoregional recurrence‐free survival (HR = 1.87, 95% CI 1.03–3.40; P = 0.03641) and recurrence‐free survival rates (HR = 1.84, 95% CI 1.01–3.34; P = 0.04206; Fig. 2B).

Figure 2.

Figure 2

Validation of the extracted four‐gene classifier in an independent HNSCC cohort. Kaplan–Meier curves for the endpoints overall (A), locoregional recurrence‐free and recurrence‐free survival (B) for patients stratified according to the four‐gene classifier of the adjuvant radio(chemo)therapy‐treated LMU‐KKG HNSCC cohort. Survival curves are shown for all cases (left panel) and for the HPV‐negative cases (right panel). Hazard ratios and median survival times with 95% contingency intervals and P‐values were calculated by Log‐rank test. Heatmap of mRNA expression levels of the four signature genes arranged according to risk scores (A, lower panel) for patients of the adjuvant radio(chemo)therapy‐treated LMU‐KKG cohort. All genes show a tendency toward higher expression in patients of the high‐risk group. NE, not estimable.

In order to assess whether the classifier was an independent prognostic factor, associations of known clinicopathological factors with the high‐ and low‐risk groups were tested. HPV status was associated with the signature‐defined risk groups of the TCGA training and validation set (Table 1). All other demographic parameters were equally distributed among the risk groups in both TCGA subsets and the LMU‐KKG HNSCC cohort. Univariate Cox regression analyses revealed a significant association of HPV status with OS in both cohorts, which was also reflected by an improved OS of oropharyngeal tumors (n = 7; all HPV‐positive) compared to tumors of the oral cavity in the TCGA sets (Table S4).

Due to the lack of the presence of HPV‐positive cases in the TCGA high‐risk groups, it was unfeasible to conduct a multivariate analysis. Instead, survival analyses were carried out stratified to HPV‐negative patients demonstrating a trend toward worse survival of high‐risk patients despite small group sizes in the TCGA subsets (Fig. 1). An additional survival analysis performed in the pooled HPV‐negative tumors of the TCGA subsets (training and validation) demonstrated a significant separation of the four‐gene signature classified risk groups low‐risk and high‐risk (HR 2.60, 95% CI 1.08–6.26; P = 0.001444) (Fig. S2). Inclusion of HPV status in a multivariate Cox‐proportional hazard model in the LMU‐KKG cohort revealed a significant contribution of the four‐gene classifier and HPV status to the prediction model (Table 2). The inclusion of both parameters HPV status and the four‐gene signature in the model was justified because there was no significant association between them (Fisher's exact test P‐value = 0.6406; Table 1). An additional stratified model including an interaction term revealed no significant interaction between the four‐gene signature and HPV status (P = 0.73). However, due to the low number of events in the group of HPV‐positive cases (4/23 events; 17% of patients), the results cannot consider being meaningful. Instead, we performed survival analyses considering HPV‐negative cases only (Fig. 2).

Table 2.

Multivariate Cox regression analysis of the four‐gene classifier and HPV status with OS in the LMU‐KKG HNSCC cohort

Cohort Parameter No. of cases HR 95% CI HR P‐value (univariate analysis) P‐value
LMU‐KKG Four‐gene classifier
(high‐risk vs low‐risk)
55 vs 53 2.17 1.18‐4.00 0.022 0.013
+
HPV status
(negative vs positive)
85 vs 23 4.04 1.44‐11.30 0.0067 0.0078

Next, we assessed the association of DNA gains of 16q24.3 with mRNA expression levels of the signature genes and the four‐gene classifier risk scores. In both TCGA subsets, significantly higher (P < 0.05) risk scores were observed for HNSCC cases with compared to cases without DNA gains of 16q24.3, also when stratified to HPV‐negative patients (Fig. 3). This was also the case on single gene level (Fig. S3).

Figure 3.

Figure 3

DNA gains of chromosomal band 16q24.3 are associated with increased risk scores in radio(chemo)therapy‐treated patients of the TCGA HNSCC training and validation set. Distribution of risk scores in all (left panel) and in HPV‐negative (right panel) cases with gain and without gain of the chromosomal band 16q24.3 in the training (all cases: n = 40, HPV‐negative cases: n = 31) (A) and validation (all cases: n = 59, HPV‐negative cases: n = 47) (B) set within the subgroup of radio(chemo)therapy‐treated HNSCC of the TCGA cohort. Patients with a DNA gain of 16q24.3 had significantly higher risk scores compared to patients without a DNA gain of 16q24.3 (Mann–Whitney U‐test). The association remained after stratification to HPV‐negative patients. The results are in whole based upon data generated by the TCGA Research Network: http://cancergenome.nih.gov (Cancer Genome Atlas, 2015; Cerami et al., 2012; Gao et al., 2013).

To get insights into the biological functions of the signature genes, a GSEA of gene expression data with regard to the signature‐defined high‐ versus low‐risk groups was performed. Twenty‐nine significantly (FDR < 0.05) up‐regulated (Table S5) and 53 down‐regulated gene sets (Table S6), respectively, in the high‐risk group compared to the low‐risk group were identified. Several significantly up‐regulated gene sets were related to FGFR signaling. Further, we found up‐regulated gene sets associated with PI3K cascade, PD1 signaling, and TCR signaling. Most of the down‐regulated gene sets were associated with the regulation of cell cycle, apoptosis, and DNA damage response.

In addition, a GAN was reconstructed. The first neighborhood (direct neighbors, n = 92) and second neighborhood (neighbors of first neighbors, n = 2972) of the four signature genes were extracted, including the information on the correlation of genes (Tables S7 and S8). Interestingly, direct network connections for three (APRT, CENPBD1, CHMP1A) of the four signature genes were observed, whereas an indirect connection was detected for galactosamine (N‐acetyl)‐6‐sulfatase (GALNS) (Fig. 4). All 50% of first neighborhood genes are localized on chromosome 16q, and a significant overrepresentation of 16q genes was observed within the first neighborhood compared to the entire network (Fisher's exact test P < 0.001).

Figure 4.

Figure 4

First neighborhood of the four signature genes from the reconstructed GAN in the TCGA HNSCC cohort. First neighborhood network of the four signature genes (red) extracted from the GAN. De novo network reconstruction was based on partial correlation of differentially expressed genes between high‐risk and low‐risk group patients in the TCGA HNSCC cohort (n = 98) using the GeneNet method. Black lines (edges) represent positive, and blue lines negative correlations (interactions) between genes. Dotted lines indicate the connection between the first neighborhood genes of GALNS and the first neighborhood genes of the other three signature genes. Genes, apart from the four signature genes, localized on 16q are colored in cyan.

Subsequent pathway enrichment analysis of genes from the first and second neighborhoods of the four signature genes revealed 493 significantly enriched pathways (FDR < 0.05) (Table S9). The top 50 identified pathways ordered according to the smallest FDR included mainly pathways associated with cell cycle, apoptosis, cell adhesion, immune response, JAK‐STAT‐signaling, signaling by SCF‐KIT, EGFR, ERBB, WNT, mTOR, and PIP3/AKT signaling.

Integration of copy number data and transcriptome data revealed a significant association (FDR < 0.1) of the genomic copy number status and the corresponding mRNA expression for 2952 out of the 7755 genes preselected for GAN reconstruction, including 64 out of 92 first neighborhood signature genes.

This represents a significant enrichment of genes within the first neighborhood with a significant association of the DNA status and their mRNA expression compared to the preselection (Fisher's exact test P < 0.001). Out of the 64 genes, 46 are located on chromosome 16q and thereby show a significant overrepresentation of 16q genes with significant copy‐number‐mRNA association compared to the preselection (Table S10).

4. Discussion

Gains of chromosomal band 16q24.3 were repeatedly shown to be associated with impaired clinical outcome of HNSCC patients after radiotherapy (Bauer et al., 2008; Hess et al., 2017). Here, we investigated the impact of 16q24.3 on mRNA expression level with regard to patients’ outcome following radio(chemo)therapy. We succeeded in the discovery and validation of a prognostic four‐gene classifier, consisting of the genes APRT, CENPBD1, CHMP1A, and GALNS located on 16q24.3. In our opinion, the fact that the signature works in both cohorts (TCGA and LMU‐KKG cohort) although exhibiting significant demographic differences underlines its robustness, which is one of the most important features of clinically applicable biomarkers.

Up to now, HPV status is the most valid and robust prognostic marker in HNSCC, whereas HPV‐positive tumors are now widely regarded as a distinct clinical entity with a different molecular pathogenesis (O'Sullivan et al., 2016). This emphasizes the importance for excluding possible confounding effects of HPV status and of other clinical and pathological parameters in prognostic marker research. For this reason, the independence from other clinical parameters is a particular strength of the presented classifier and allows statistically significant stratification of patient risk groups in the subgroup of HPV‐negative HNSCC.

Smoking and alcohol abuse are established risk factors for HNSCC, also contributing to a high prevalence of multiple comorbidities (e.g., cardiovascular, pulmonary, or hepatic diseases), which have a particularly high impact on the OS of HNSCC patients (Datema et al., 2010). Consideration of tumor‐specific clinical endpoints provides a more solid basis in prognostic marker development for HNSCC. In this sense, the four‐gene classifier was also prognostic for locoregional recurrence‐free survival and recurrence‐free survival of HNSCC patients following adjuvant radio(chemo)therapy substantiating its clinical value. These results correspond to the previously reported association of DNA gains of 16q24.3 with locoregional recurrence‐free survival in HNSCC (Bauer et al., 2008). The prognostic value of the signature was additionally underlined by the significant correlation of DNA gains 16q24.3 with elevated mRNA expression levels of the four classifier genes. This is important to note because copy number alterations not necessarily lead to aberrant mRNA expression of genes (Gollin, 2014; Jarvinen et al., 2006).

An association of genomic gains on 16q24.3 with increased mRNA expression levels was previously also observed for the DNA repair‐related gene FancA, located within this chromosomal band. Further, silencing of FancA expression in HNSCC cell lines with genomic gains on 16q24.3 resulted in significantly impaired clonogenic survival upon irradiation, whereas overexpression of FancA conferred increased survival (Hess et al., 2017). Interestingly, the FancA gene was not included in the best performing model when developing a low‐complexity prognostic mRNA expression signature including 16q24.3 genes. In this regard, we have indications from another project that specific transcript isoforms of FancA are associated with HNSCC patient prognosis.

The potential exploitation of the four‐gene signature as a novel therapeutic target is dependent on their biological functions and involved pathways. A first insight into the possible functional role of the signature genes was gained from the published literature, but only little information on the genes in context with HNSCC or even with cancer was available. CHMP1A (charged multivesicular body protein 1A) is known to act as a tumor suppressor in pancreatic (Li et al., 2008, 2009) and renal cancer (You et al., 2012) by inhibition of tumor cell proliferation. However, an up‐regulation of CHMP1A in HNSCC might also have a converse effect as multifunctional roles were shown for several tumor‐associated genes (Radin and Patel, 2017). Furthermore, CENPBD1 (CENPB DNA‐binding domain containing 1) plays a role in centromere formation and could, therefore, also have an influence on the cell cycle. The enzyme APRT (adenine phosphoribosyltransferase) is involved in purine metabolism (Kamatani et al., 1984), and deficiency of the lysosomal exohydrolase GALNS is associated with the autosomal recessive disorder Mucopolysaccharidosis IV A (Horwitz and Dorfman, 1978; Matalon et al., 1974). So far, APRT and GALNS have not been described in connection with cancer or cancer‐associated molecular pathways. However, a GSEA and the investigation of our reconstructed GAN revealed evidence for specific involvement of the four signature genes in pathways associated with poor clinical outcome and therapy resistance in HNSCC: for example, the EGFR/PIP3K/AKT/mTOR pathway, involved in numerous cancer‐related processes including cell cycle progression and apoptosis, is frequently altered in HNSCC and linked to therapeutic failure (Freudlsperger et al., 2011; Niehr et al., 2018). The mTOR pathway leads to the activation of the DNA‐repair machinery, thereby inducing higher radioresistance of HNSCC cells (Bose et al., 2013). Also the identified aberrant DNA damage response could indicate the involvement of the four signature genes in the radiation resistance of tumor cells (Hosoya and Miyagawa, 2014). Moreover, cell adhesion molecules like integrins are promising targets to overcome therapeutic resistance of HNSCC cells, especially in combination with radiation and simultaneous targeting of EGFR (Eke et al., 2015). Further identified pathways included the JAK‐STAT signaling, regulating cell proliferation, cell survival, and angiogenesis (Bose et al., 2013). In combination with aberrant Interleukin and GM‐CSF signaling, this causes markedly immune system evasion of tumors, limiting the efficacy of conventional therapies (Bose et al., 2013; Wu et al., 2015). Also the reactivation of developmental processes initiated by stem cell factor (SCF)/KIT, and Wnt signaling may play a role in the therapeutic response of HNSCC tumor cells (Ischenko et al., 2008). Taken together, the involvement of the four signature genes in those pathways could explain the worse clinical outcome of high‐risk patients as defined by the four‐gene classifier. Whether the functionality of the signature is related to a radiation‐resistant phenotype, which is partly suggested by the signaling pathways discussed above, or rather to tumor aggressiveness, must be investigated in future in vitro studies.

Moreover, since copy number alterations not necessarily lead to aberrant mRNA expression of genes (Gollin, 2014; Jarvinen et al., 2006), it was an important finding for us that in the case of 16q, where the signature genes are located, the gene expression levels are significantly associated with the corresponding DNA copy number status. It underlines the prognostic value of the signature based on the previous study on a DNA gain of 16q24.3 (Bauer et al., 2008; Hess et al., 2017) and its association with compromised therapy response. Furthermore, these results in combination with the analysis of the reconstructed GAN indicate a possible cluster of co‐expressed genes on chromosome 16q, including the four signature genes, with an impact on cancer‐related processes in HNSCC. Such clusters of co‐expressed genes were previously described in the published literature (Caron et al., 2001) and are known to consist of genes involved in the same functional pathways (Lee and Sonnhammer, 2003). Based on these facts, the cluster of co‐expressed genes on 16q could be involved in poor clinical outcome of HNSCC.

5. Conclusion

In conclusion, our findings do not only provide a prognostic tool for stratification of HNSCC patients treated with adjuvant radio(chemo)therapy into groups of favorable and poor prognosis independent of other clinical parameters. They also might help identifying targets for molecular therapies, since the four‐gene signature seems to be part of a functional gene expression cluster involved in HNSCC‐associated pathways.

Author contributions

JH and KU designed the study. JH and LW designed the experiments. LW carried out the experiments. KU, MS, LW, and JH performed analysis and interpretation of the data. CM, LS, AW, CW, PB, TK, CB, and UG established the LMU‐KKG cohort including collection of clinical data and samples. JH, LW, KU, MS, and HZ interpreted the results and drafted/revised the manuscript. All authors provided critical feedback, reviewed the manuscript, and approved the final version.

Supporting information

Fig. S1. Schematic workflow for the development of the classifier and the reconstruction of the gene association network with subsequent analyses.

Fig. S2. Kaplan–Meier curves for the endpoint overall survival for the pooled HPV‐negative patients of the radio(chemo)therapy‐treated TCGA training and validation set stratified according to the four‐gene classifier.

Fig. S3. DNA gains of chromosomal band 16q24.3 are associated with increased mRNA expression levels of the signature genes in radio(chemo)therapy‐treated patients of the TCGA HNSCC cohort.

Table S1. REporting recommendations for tumour MARKer prognostic studies (REMARK) according to McShane et al. (2005).

Table S2. Patient subset of 99 radio(chemo)therapy‐treated HNSCC patients of the TCGA cohort.

Table S3. Differences of demographic and histopathological parameters between the TCGA cohort and the LMU‐KKG cohort.

Table S4. Univariate Cox regression analyses of clinicopathological parameters and the four‐gene classifier with overall survival in the TCGA HNSCC training and validation set and the LMU‐KKG HNSCC cohort.

Table S5. Gene set enrichment analysis (GSEA): Significantly (FDR < 0.05) up‐regulated sets of genes between the four‐gene signature high and low‐risk groups of the TCGA cohort.

Table S6. Gene set enrichment analysis (GSEA): Significantly (FDR < 0.05) down‐regulated sets of genes between the four‐gene signature high and low‐risk groups of the TCGA cohort.

Table S7. Correlation of the first neighborhood genes of the reconstructed gene association network.

Table S8. Correlation of the second neighborhood genes of the reconstructed gene association network.

Table S9. Significantly enriched pathways (FDR < 0.05) of first and second neighborhood genes of the four signature genes from the reconstructed GAN.

Table S10. Significant results of the sigaR cis‐test.

Acknowledgements

This study was supported by the iMed project on ‘Multi‐Scale Integrative Biology of HNSCC—Molecular Consequences of the Metabolic State and HPV‐Infection Status on Radiotherapy Response’. The authors want to thank L. Dajka, C. Innerlohinger, U. Pflugradt, C.‐M. Pflüger, and U. Buchholz for excellent technical assistance, all co‐workers of the Clinical Cooperation Group ‘Personalized Radiotherapy in Head and Neck Cancer’ for scientific support. The results shown here are in part based upon data generated by the TCGA Research Network: http://cancergenome.nih.gov/.

Kristian Unger and Julia Hess are equally contributing senior authors.

References

  1. Ang KK, Harris J, Wheeler R, Weber R, Rosenthal DI, Nguyen‐Tan PF, Westra WH, Chung CH, Jordan RC, Lu C et al (2010) Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med 363, 24–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Bauer VL, Braselmann H, Henke M, Mattern D, Walch A, Unger K, Baudis M, Lassmann S, Huber R, Wienberg J et al (2008) Chromosomal changes characterize head and neck cancer with poor prognosis. J Mol Med (Berl) 86, 1353–1365. [DOI] [PubMed] [Google Scholar]
  3. Bose P, Brockton NT and Dort JC (2013) Head and neck cancer: from anatomy to biology. Int J Cancer 133, 2013–2023. [DOI] [PubMed] [Google Scholar]
  4. Cancer Genome Atlas, N. (2015) Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 517, 576–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Caron H, van Schaik B, van der Mee M, Baas F, Riggins G, van Sluis P, Hermus MC, van Asperen R, Boon K, Voute PA et al (2001) The human transcriptome map: clustering of highly expressed genes in chromosomal domains. Science 291, 1289–1292. [DOI] [PubMed] [Google Scholar]
  6. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E et al (2012) The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2, 401–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Cho H, Yu A, Kim S, Kang J and Hong S‐M (2009) Robust likelihood‐based survival modeling with microarray data. J Stat Softw 29, 16. [Google Scholar]
  8. Chung CH, Parker JS, Karaca G, Wu J, Funkhouser WK, Moore D, Butterfoss D, Xiang D, Zanation A, Yin X et al (2004) Molecular classification of head and neck squamous cell carcinomas using patterns of gene expression. Cancer Cell 5, 489–500. [DOI] [PubMed] [Google Scholar]
  9. Datema FR, Ferrier MB, van der Schroeff MP and Baatenburg de Jong RJ (2010) Impact of comorbidity on short‐term mortality and overall survival of head and neck cancer patients. Head Neck 32, 728–736. [DOI] [PubMed] [Google Scholar]
  10. Eke I, Zscheppang K, Dickreuter E, Hickmann L, Mazzeo E, Unger K, Krause M, Cordes N (2015) Simultaneous beta1 integrin‐EGFR targeting and radiosensitization of human head and neck cancer. J Natl Cancer Inst 107, pii: dju419. [DOI] [PubMed] [Google Scholar]
  11. Eustace A, Mani N, Span PN, Irlam JJ, Taylor J, Betts GN, Denley H, Miller CJ, Homer JJ, Rojas AM et al (2013) A 26‐gene hypoxia signature predicts benefit from hypoxia‐modifying therapy in laryngeal cancer but not bladder cancer. Clin Cancer Res 19, 4879–4888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Freudlsperger C, Burnett JR, Friedman JA, Kannabiran VR, Chen Z and Van Waes C (2011) EGFR‐PI3K‐AKT‐mTOR signaling in head and neck squamous cell carcinomas: attractive targets for molecular‐oriented therapy. Expert Opin Ther Targets 15, 63–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E et al (2013) Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6, pl1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Ginos MA, Page GP, Michalowicz BS, Patel KJ, Volker SE, Pambuccian SE, Ondrey FG, Adams GL and Gaffney PM (2004) Identification of a gene expression signature associated with recurrent disease in squamous cell carcinoma of the head and neck. Cancer Res 64, 55–63. [DOI] [PubMed] [Google Scholar]
  15. Gollin SM (2014) Cytogenetic alterations and their molecular genetic correlates in head and neck squamous cell carcinoma: a next generation window to the biology of disease. Genes Chromosom Cancer 53, 972–990. [DOI] [PubMed] [Google Scholar]
  16. Hess J, Unger K, Orth M, Schotz U, Schuttrumpf L, Zangen V, Gimenez‐Aznar I, Michna A, Schneider L, Stamp R et al (2017) Genomic amplification of Fanconi anemia complementation group A (FancA) in head and neck squamous cell carcinoma (HNSCC): Cellular mechanisms of radioresistance and clinical relevance. Cancer Lett 386, 87–99. [DOI] [PubMed] [Google Scholar]
  17. Hesselink AT, van den Brule AJ, Groothuismink ZM, Molano M, Berkhof J, Meijer CJ and Snijders PJ (2005) Comparison of three different PCR methods for quantifying human papillomavirus type 16 DNA in cervical scrape specimens. J Clin Microbiol 43, 4868–4871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Horwitz AL and Dorfman A (1978) The enzymic defect in Morquio's disease: the specificity of N‐acetylhexosamine sulfatases. Biochem Biophys Res Commun 80, 819–825. [DOI] [PubMed] [Google Scholar]
  19. Hosoya N and Miyagawa K (2014) Targeting DNA damage response in cancer therapy. Cancer Sci 105, 370–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Ischenko I, Seeliger H, Schaffer M, Jauch KW and Bruns CJ (2008) Cancer stem cells: how can we target them? Curr Med Chem 15, 3171–3184. [DOI] [PubMed] [Google Scholar]
  21. Jarvinen AK, Autio R, Haapa‐Paananen S, Wolf M, Saarela M, Grenman R, Leivo I, Kallioniemi O, Makitie AA and Monni O (2006) Identification of target genes in laryngeal squamous cell carcinoma by high‐resolution copy number and gene expression microarray analyses. Oncogene 25, 6997–7008. [DOI] [PubMed] [Google Scholar]
  22. Kamatani N, Kubota M, Willis EH, Frincke LA, Carson DA (1984) 5’‐Methylthioadenosine is the major source of adenine in human cells. Adv Exp Med Biol 165 (Pt B), 83–88. [DOI] [PubMed] [Google Scholar]
  23. Keck MK, Zuo Z, Khattri A, Stricker TP, Brown CD, Imanguli M, Rieke D, Endhardt K, Fang P, Bragelmann J et al (2015) Integrative analysis of head and neck cancer identifies two biologically distinct HPV and three non‐HPV subtypes. Clin Cancer Res 21, 870–881. [DOI] [PubMed] [Google Scholar]
  24. Lee JM and Sonnhammer EL (2003) Genomic gene clustering analysis of pathways in eukaryotes. Genome Res 13, 875–882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Leemans CR, Braakhuis BJ and Brakenhoff RH (2011) The molecular biology of head and neck cancer. Nat Rev Cancer 11, 9–22. [DOI] [PubMed] [Google Scholar]
  26. Li J, Belogortseva N, Porter D and Park M (2008) Chmp1A functions as a novel tumor suppressor gene in human embryonic kidney and ductal pancreatic tumor cells. Cell Cycle 7, 2886–2893. [DOI] [PubMed] [Google Scholar]
  27. Li J, Orr B, White K, Belogortseva N, Niles R, Boskovic G, Nguyen H, Dykes A and Park M (2009) Chmp 1A is a mediator of the anti‐proliferative effects of all‐trans retinoic acid in human pancreatic cancer cells. Mol Cancer 8, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Lian M, Fang J, Han D, Ma H, Feng L, Wang R and Yang F (2013) Microarray gene expression analysis of tumorigenesis and regional lymph node metastasis in laryngeal squamous cell carcinoma. PLoS One 8, e84854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Lindh M, Gorander S, Andersson E, Horal P, Mattsby‐Balzer I and Ryd W (2007) Real‐time Taqman PCR targeting 14 human papilloma virus types. J Clin Virol 40, 321–324. [DOI] [PubMed] [Google Scholar]
  30. Lohavanichbutr P, Mendez E, Holsinger FC, Rue TC, Zhang Y, Houck J, Upton MP, Futran N, Schwartz SM, Wang P et al (2013) A 13‐gene signature prognostic of HPV‐negative OSCC: discovery and external validation. Clin Cancer Res 19, 1197–1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Love MI, Huber W and Anders S (2014) Moderated estimation of fold change and dispersion for RNA‐seq data with DESeq2. Genome Biol 15, 550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Maihoefer C, Schüttrumpf L, Macht C, Pflugradt U, Hess J, Schneider L, Woischke C, Walch A, Baumeister P, Kirchner T et al (2018) Postoperative (chemo) radiation in patients with squamous cell cancers of the head and neck – clinical results from the cohort of the clinical cooperation group “Personalized Radiotherapy in Head and Neck Cancer”. Radiat Oncol 13, 123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Marcu LG and Yeoh E (2009) A review of risk factors and genetic alterations in head and neck carcinogenesis and implications for current and future approaches to treatment. J Cancer Res Clin Oncol 135, 1303–1314. [DOI] [PubMed] [Google Scholar]
  34. Matalon R, Arbogast B, Justice P, Brandt IK and Dorfman A (1974) Morquio's syndrome: deficiency of a chondroitin sulfate N‐acetylhexosamine sulfate sulfatase. Biochem Biophys Res Commun 61, 759–765. [DOI] [PubMed] [Google Scholar]
  35. McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM, Statistics Subcommittee of the N.C.I.E.W.G.o.C.D. (2005) REporting recommendations for tumor MARKer prognostic studies (REMARK). Nat Clin Pract Urol 2, 416–422. [PubMed] [Google Scholar]
  36. Mirghani H, Amen F, Blanchard P, Moreau F, Guigay J, Hartl DM, Lacau St Guily J (2015) Treatment de‐escalation in HPV‐positive oropharyngeal carcinoma: ongoing trials, critical issues and perspectives. Int J Cancer 136, 1494–1503. [DOI] [PubMed] [Google Scholar]
  37. Niehr F, Eder T, Pilz T, Konschak R, Treue D, Klauschen F, Bockmayr M, Turkmen S, Johrens K, Budach V et al (2018) Multilayered omics‐based analysis of a head and neck cancer model of cisplatin resistance reveals intratumoral heterogeneity and treatment‐induced clonal selection. Clin Cancer Res 24, 158–168. [DOI] [PubMed] [Google Scholar]
  38. Opgen‐Rhein R and Strimmer K (2007) From correlation to causation networks: a simple approximate learning algorithm and its application to high‐dimensional plant gene expression data. BMC Syst Biol 1, 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. O'Sullivan B, Huang SH, Siu LL, Waldron J, Zhao H, Perez‐Ordonez B, Weinreb I, Kim J, Ringash J, Bayley A et al (2013) Deintensification candidate subgroups in human papillomavirus‐related oropharyngeal cancer according to minimal risk of distant metastasis. J Clin Oncol 31, 543–550. [DOI] [PubMed] [Google Scholar]
  40. O'Sullivan B, Huang SH, Su J, Garden AS, Sturgis EM, Dahlstrom K, Lee N, Riaz N, Pei X, Koyfman SA et al (2016) Development and validation of a staging system for HPV‐related oropharyngeal cancer by the International Collaboration on Oropharyngeal cancer Network for Staging (ICON‐S): a multicentre cohort study. Lancet Oncol 17, 440–451. [DOI] [PubMed] [Google Scholar]
  41. Patro R, Duggal G, Love MI, Irizarry RA and Kingsford C (2017) Salmon provides fast and bias‐aware quantification of transcript expression. Nat Methods 14, 417–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Pavon MA, Parreno M, Tellez‐Gabriel M, Sancho FJ, Lopez M, Cespedes MV, Casanova I, Lopez‐Pousa A, Mangues MA, Quer M et al (2012) Gene expression signatures and molecular markers associated with clinical outcome in locally advanced head and neck carcinoma. Carcinogenesis 33, 1707–1716. [DOI] [PubMed] [Google Scholar]
  43. Radin DP and Patel P (2017) BDNF: an oncogene or tumor suppressor? Anticancer Res 37, 3983–3990. [DOI] [PubMed] [Google Scholar]
  44. Rocco JW, Leong CO, Kuperwasser N, DeYoung MP and Ellisen LW (2006) p63 mediates survival in squamous cell carcinoma by suppression of p73‐dependent apoptosis. Cancer Cell 9, 45–56. [DOI] [PubMed] [Google Scholar]
  45. Roepman P, de Koning E, van Leenen D, de Weger RA, Kummer JA, Slootweg PJ and Holstege FC (2006) Dissection of a metastatic gene expression signature into distinct components. Genome Biol 7, R117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T (2003) Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13, 2498–2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Simon RM, Paik S and Hayes DF (2009) Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst 101, 1446–1452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Slebos RJ, Yi Y, Ely K, Carter J, Evjen A, Zhang X, Shyr Y, Murphy BM, Cmelak AJ, Burkey BB et al (2006) Gene expression differences associated with human papillomavirus status in head and neck squamous cell carcinoma. Clin Cancer Res 12, 701–709. [DOI] [PubMed] [Google Scholar]
  49. Smeets SJ, Hesselink AT, Speel EJ, Haesevoets A, Snijders PJ, Pawlita M, Meijer CJ, Braakhuis BJ, Leemans CR and Brakenhoff RH (2007) A novel algorithm for reliable detection of human papillomavirus in paraffin embedded head and neck cancer specimen. Int J Cancer 121, 2465–2472. [DOI] [PubMed] [Google Scholar]
  50. Tawk B, Schwager C, Deffaa O, Dyckhoff G, Warta R, Linge A, Krause M, Weichert W, Baumann M, Herold‐Mende C et al (2016) Comparative analysis of transcriptomics based hypoxia signatures in head‐ and neck squamous cell carcinoma. Radiother Oncol 118, 350–358. [DOI] [PubMed] [Google Scholar]
  51. Toustrup K, Sorensen BS, Alsner J and Overgaard J (2012) Hypoxia gene expression signatures as prognostic and predictive markers in head and neck radiotherapy. Semin Radiat Oncol 22, 119–127. [DOI] [PubMed] [Google Scholar]
  52. van de Wiel MA, Kim KI, Vosse SJ, van Wieringen WN, Wilting SM and Ylstra B (2007) CGHcall: calling aberrations for array CGH tumor profiles. Bioinformatics 23, 892–894. [DOI] [PubMed] [Google Scholar]
  53. van Wieringen WN, Unger K, Leday GG, Krijgsman O, de Menezes RX, Ylstra B and van de Wiel MA (2012) Matching of array CGH and gene expression microarray features for the purpose of integrative genomic analyses. BMC Bioinformatics 13, 80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. van Wieringen WN and van de Wiel MA (2009) Nonparametric testing for DNA copy number induced differential mRNA gene expression. Biometrics 65, 19–29. [DOI] [PubMed] [Google Scholar]
  55. Wood O, Woo J, Seumois G, Savelyeva N, McCann KJ, Singh D, Jones T, Peel L, Breen MS, Ward M et al (2016) Gene expression analysis of TIL rich HPV‐driven head and neck tumors reveals a distinct B‐cell signature when compared to HPV independent tumors. Oncotarget 7, 56781–56797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Wu AA, Drake V, Huang HS, Chiu S and Zheng L (2015) Reprogramming the tumor microenvironment: tumor‐induced immunosuppressive factors paralyze T cells. Oncoimmunology 4, e1016700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Wu G, Feng X and Stein L (2010) A human functional protein interaction network and its application to cancer data analysis. Genome Biol 11, R53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. You Z, Xin Y, Liu Y, Sun J, Zhou G, Gao H, Xu P, Chen Y, Chen G, Zhang L et al (2012) Chmp1A acts as a tumor suppressor gene that inhibits proliferation of renal cell carcinoma. Cancer Lett 319, 190–196. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Fig. S1. Schematic workflow for the development of the classifier and the reconstruction of the gene association network with subsequent analyses.

Fig. S2. Kaplan–Meier curves for the endpoint overall survival for the pooled HPV‐negative patients of the radio(chemo)therapy‐treated TCGA training and validation set stratified according to the four‐gene classifier.

Fig. S3. DNA gains of chromosomal band 16q24.3 are associated with increased mRNA expression levels of the signature genes in radio(chemo)therapy‐treated patients of the TCGA HNSCC cohort.

Table S1. REporting recommendations for tumour MARKer prognostic studies (REMARK) according to McShane et al. (2005).

Table S2. Patient subset of 99 radio(chemo)therapy‐treated HNSCC patients of the TCGA cohort.

Table S3. Differences of demographic and histopathological parameters between the TCGA cohort and the LMU‐KKG cohort.

Table S4. Univariate Cox regression analyses of clinicopathological parameters and the four‐gene classifier with overall survival in the TCGA HNSCC training and validation set and the LMU‐KKG HNSCC cohort.

Table S5. Gene set enrichment analysis (GSEA): Significantly (FDR < 0.05) up‐regulated sets of genes between the four‐gene signature high and low‐risk groups of the TCGA cohort.

Table S6. Gene set enrichment analysis (GSEA): Significantly (FDR < 0.05) down‐regulated sets of genes between the four‐gene signature high and low‐risk groups of the TCGA cohort.

Table S7. Correlation of the first neighborhood genes of the reconstructed gene association network.

Table S8. Correlation of the second neighborhood genes of the reconstructed gene association network.

Table S9. Significantly enriched pathways (FDR < 0.05) of first and second neighborhood genes of the four signature genes from the reconstructed GAN.

Table S10. Significant results of the sigaR cis‐test.


Articles from Molecular Oncology are provided here courtesy of Wiley

RESOURCES