There is an unmet need for effective targeted therapies for patients with advanced head and neck squamous cell carcinoma (HNSCC). We correlated gene expression, gene copy numbers, and point mutations in 45 human papillomavirus–negative HNSCC cell lines with the sensitivity to 220 anticancer drugs to discover predictive associations to genetic alterations. The drug response profiles revealed diverse efficacy of the tested drugs across the cell lines. Several genomic abnormalities and gene expression differences were associated with response to mTOR, MEK, and EGFR inhibitors. NOTCH1 and FAT1 were the most commonly mutated genes after TP53 and also showed some association with response to MEK and/or EGFR inhibitors. MYC amplification and FAM83H overexpression associated with sensitivity to EGFR inhibitors, and PTPRD deletion with poor sensitivity to MEK inhibitors. The connection between high FAM83H expression and responsiveness to the EGFR inhibitor erlotinib was validated by gene silencing and from the data set at the Cancer Cell Line Encyclopedia. The data provide several novel genomic alterations that associated to the efficacy of targeted drugs in HNSCC. These findings require further validation in experimental models and clinical series. Mol Cancer Ther; 17(9); 2060–71. ©2018 AACR.

Head and neck squamous cell carcinoma (HNSCC) is a common type of human cancer worldwide (1). Local HNSCCs are often treated with surgery, radiotherapy, and/or chemoradiotherapy, but cancer recurrence is frequent, and emergence of distant metastases invariably leads to death (2, 3). Systemic treatments for advanced HNSCC often consist of chemotherapeutic drugs, such as platinum salts and taxanes, but these drugs have only moderate efficacy (4). The only approved targeted drugs for advanced HNSCC are cetuximab, a monoclonal antibody that binds to the epidermal growth factor receptor (EGFR), and, recently, the anti-PD-1 antibodies nivolumab and pembrolizumab (5). Despite EGFR is frequently amplified and overexpressed in HNSCC, cetuximab has shown only moderate efficacy (6, 7), and there are no clinically approved predictive biomarkers for cetuximab response. There is a clear unmet need for novel, effective targeted treatments and accompanying biomarkers for efficacy.

Recent comprehensive genomic analyses of HNSCCs have greatly advanced our understanding of the disease and the key molecular alterations that drive these cancers (8–13). However, the translation of the molecular aberrations into treatment strategies for patients is challenging as the efficacy of many anticancer drugs is influenced by the genomic and transcriptomic heterogeneity of tumors (14). Numerous novel anticancer drugs are entering the clinical phase of testing, and testing all of them and their potential combinations in clinical trials is challenging. Combining molecular and genetic analyses with comprehensive preclinical compound screening in vitro might reveal new drug targets, and could facilitate rational drug testing in clinical trials.

HNSCCs comprise both human papillomavirus (HPV)-positive and HPV-negative cancers, the latter being more common. Patients with HPV-negative cancer have, in general, less favorable survival as compared with those with HPV-positive HNSCC (15). Recurrent or metastatic HPV-positive cancers resemble genetically HPV-negative tumors (16). In the current study, we performed a large-scale drug screen on 45 HPV-negative HNSCC cell lines, and integrated the compound sensitivity landscapes with detailed genomic and gene expression analyses to identify novel drugs that might be effective for HNSCC along with predictive biomarkers for these drugs.

Cell lines

The series consists of 45 HPV-negative HNSCC cell lines established from HNSCC (Table 1). The cell lines were established at the Department of Otorhinolaryngology-Head and Neck Surgery, Turku University Hospital (Turku, Finland) as described elsewhere (17, 18). Thirty-one (69%) of the 45 patients whose cancer gave rise to a cell line were male, and the mean age was 62 years at the time of tissue sampling. A majority (36 of 45, 80%) of the cell lines originated from a primary tumor, 4 (9%) from a primary tumor that persisted after treatment, and 5 (11%) from recurrent cancer. Most of the cell lines (n = 33, 73%) originated from an oral cavity or oropharyngeal tumor, 11 (24%) from larynx cancers, and 1 (2%) from a maxillary sinus cancer. All cell lines tested negative for mycoplasma before the experiments using a VenorGeM Mycoplasma PCR Detection Kit (Minerva Biolabs GmbH).

Table 1.

Clinical and pathologic characteristics of the HNSCC cell lines

Cell lineSexaAgebTNMSpecimen siteTypecGradePassage
UT-SCC-2 60 T4N1M0 Baseos oris pri G2 p10 
UT-SCC-6A 51 T2N1M0 Larynx rec G1 p31 
UT-SCC-8 42 T2N0M0 Larynx pri G1 p35 
UT-SCC-10 62 T1N0M0 Tongue pri G2 p6–7 
UT-SCC-11 58 T1N0M0 Larynx rec G2 p18 
UT-SCC-14 25 T3N1M0 Tongue pri (per) G2 p9 
UT-SCC-16A 77 T3N0M0 Tongue pri G3 p23 
UT-SCC-19A 44 T4N0M0 Larynx pri G2 p15 
UT-SCC-21 79 T3N0M0 Tongue pri G2 p19 
UT-SCC-24A 41 T2N0M0 Tongue pri G2 p29 
UT-SCC-27 71 T2N0M0 Gingiva of maxilla rec G3 p10 
UT-SCC-28 58 T2N0M0 Floor of mouth pri (per) G1 p23 
UT-SCC-29 82 T2N0M0 Larynx pri G1 p17 
UT-SCC-30 77 T3N1M0 Oral tongue pri G1 p41 
UT-SCC-36 46 T4N1M0 Floor of mouth pri G3 p20 
UT-SCC-37 61 T2N0M0 Gingiva of mandib. pri G1 p25 
UT-SCC-38 66 T2N0M0 Larynx pri G2 p5 
UT-SCC-40 65 T3N0M0 Tongue pri G1 p6 
UT-SCC-42A 43 T4N3M0 Larynx pri G3 p23 
UT-SCC-43 75 T4N1M0 Gingiva of mandib. pri G2 p10 
UT-SCC-44 71 T4N2BM0 Gingiva of maxilla pri (per) G3 p24 
UT-SCC-47 78 T2N0M0 Floor of mouth pri G3 p9 
UT-SCC-49 76 T2N0M0 Larynx pri G2 p29 
UT-SCC-53 76 T4N2cM0 Maxillary sinus pri G1 p19–20 
UT-SCC-54A 58 T2N0M0 Buchal mucosa pri G1 p35 
UT-SCC-55 76 T4N1M0 Gingiva pri G2 p23–24 
UT-SCC-60A 59 T4N1M0 Tonsil (left) pri G1 p12 
UT-SCC-65 68 T4N2CM0 Soft palate and tonsilla pri G2 p7–8 
UT-SCC-67 80 T2N0M0 Tongue pri G2 p16 
UT-SCC-72 50 T4N2M0 Gingiva pri (per) G2 p17–18 
UT-SCC-73 86 T1N0M0 Tongue pri G2 p18 
UT-SCC-74A 31 T3N1M0 Tongue pri G1-G2 p44 
UT-SCC-75 56 T2N2BM0 Larynx pri G2 p30+ 
UT-SCC-76A 52 T3N0M0 Tongue pri G2 p17 
UT-SCC-81 48 T2N0M0 Tongue pri G1 p18 
UT-SCC-87 29 T3N1M0 Tongue pri G1 p31 
UT-SCC-95 83 T1N0M0 Tongue pri G1 p25 
UT-SCC-99 80 T3N0M0 Tongue pri G2 p5–6 
UT-SCC-106A 59 T1AN0M0 Larynx pri G2 p12 
UT-SCC-107 46 T4N2CM0 Larynx pri G2 p24 
UT-SCC-110A 37 T4N0M0 Maxilla rec G3 p23 
UT-SCC-112 94 T2N0M0 Gingivae mandib. pri G1 p16 
UT-SCC-114 75 T3N0M0 Lower lip pri G1 p10 
UT-SCC-122 36 T3NM0 Mucosae bucchae pri G2 p8 
UT-SCC-126A 81 T2N1M0 Lower lip rec G1 p8 
Cell lineSexaAgebTNMSpecimen siteTypecGradePassage
UT-SCC-2 60 T4N1M0 Baseos oris pri G2 p10 
UT-SCC-6A 51 T2N1M0 Larynx rec G1 p31 
UT-SCC-8 42 T2N0M0 Larynx pri G1 p35 
UT-SCC-10 62 T1N0M0 Tongue pri G2 p6–7 
UT-SCC-11 58 T1N0M0 Larynx rec G2 p18 
UT-SCC-14 25 T3N1M0 Tongue pri (per) G2 p9 
UT-SCC-16A 77 T3N0M0 Tongue pri G3 p23 
UT-SCC-19A 44 T4N0M0 Larynx pri G2 p15 
UT-SCC-21 79 T3N0M0 Tongue pri G2 p19 
UT-SCC-24A 41 T2N0M0 Tongue pri G2 p29 
UT-SCC-27 71 T2N0M0 Gingiva of maxilla rec G3 p10 
UT-SCC-28 58 T2N0M0 Floor of mouth pri (per) G1 p23 
UT-SCC-29 82 T2N0M0 Larynx pri G1 p17 
UT-SCC-30 77 T3N1M0 Oral tongue pri G1 p41 
UT-SCC-36 46 T4N1M0 Floor of mouth pri G3 p20 
UT-SCC-37 61 T2N0M0 Gingiva of mandib. pri G1 p25 
UT-SCC-38 66 T2N0M0 Larynx pri G2 p5 
UT-SCC-40 65 T3N0M0 Tongue pri G1 p6 
UT-SCC-42A 43 T4N3M0 Larynx pri G3 p23 
UT-SCC-43 75 T4N1M0 Gingiva of mandib. pri G2 p10 
UT-SCC-44 71 T4N2BM0 Gingiva of maxilla pri (per) G3 p24 
UT-SCC-47 78 T2N0M0 Floor of mouth pri G3 p9 
UT-SCC-49 76 T2N0M0 Larynx pri G2 p29 
UT-SCC-53 76 T4N2cM0 Maxillary sinus pri G1 p19–20 
UT-SCC-54A 58 T2N0M0 Buchal mucosa pri G1 p35 
UT-SCC-55 76 T4N1M0 Gingiva pri G2 p23–24 
UT-SCC-60A 59 T4N1M0 Tonsil (left) pri G1 p12 
UT-SCC-65 68 T4N2CM0 Soft palate and tonsilla pri G2 p7–8 
UT-SCC-67 80 T2N0M0 Tongue pri G2 p16 
UT-SCC-72 50 T4N2M0 Gingiva pri (per) G2 p17–18 
UT-SCC-73 86 T1N0M0 Tongue pri G2 p18 
UT-SCC-74A 31 T3N1M0 Tongue pri G1-G2 p44 
UT-SCC-75 56 T2N2BM0 Larynx pri G2 p30+ 
UT-SCC-76A 52 T3N0M0 Tongue pri G2 p17 
UT-SCC-81 48 T2N0M0 Tongue pri G1 p18 
UT-SCC-87 29 T3N1M0 Tongue pri G1 p31 
UT-SCC-95 83 T1N0M0 Tongue pri G1 p25 
UT-SCC-99 80 T3N0M0 Tongue pri G2 p5–6 
UT-SCC-106A 59 T1AN0M0 Larynx pri G2 p12 
UT-SCC-107 46 T4N2CM0 Larynx pri G2 p24 
UT-SCC-110A 37 T4N0M0 Maxilla rec G3 p23 
UT-SCC-112 94 T2N0M0 Gingivae mandib. pri G1 p16 
UT-SCC-114 75 T3N0M0 Lower lip pri G1 p10 
UT-SCC-122 36 T3NM0 Mucosae bucchae pri G2 p8 
UT-SCC-126A 81 T2N1M0 Lower lip rec G1 p8 

aM = male, F = female; bage in years; cPri, primary tumor; pri (per), primary persistent tumor; rec = recurrent tumor.

The cells were cultured in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 2 mmol/L L-glutamine, 10% fetal bovine serum, nonessential amino acids solution, penicillin (100 U/mL), and streptomycin (100 μg/mL) at 37°C in an atmosphere of 5% CO2 (all reagents were purchased from Lonza BioWhittaker). Genotyping for HPV 6, 11, 16, 18, 26, 31, 33, 35, 39, 42, 43, 44, 45, 51, 52, 53, 56, 58, 59, 66, 68, 70, 73, and 82 strains was performed using a Multiplex HPV Genotyping Kit (DiaMex) in PCR-amplified samples of genomic DNA isolated from the cell lines. All cell lines were negative for all the HPV strains tested. Normal skin fibroblasts were obtained from 3 patients whose tumors gave rise to the UT-SCC-19A, UT-SCC-29, and UT-SCC-45 cell lines, and they were cultured as the cancer cells except that 20 mmol/L HEPES (Invitrogen) was added to the cell culture media. CCD 1106 KERTr (ATCC CRL-2309) keratinocytes were grown in keratinocyte-serum free medium supplemented with epidermal growth factor and bovine pituitary extract (Invitrogen).

Nucleic acid isolation

Genomic DNA was isolated from the cell lines using a DNeasy Blood and Tissue Kit (Qiagen) according to the manufacturer's protocol. The concentration and purity of the genomic DNA was estimated with a NanoDrop spectrophotometer (Thermo Scientific). Total RNA was isolated using a Macherey-Nagel (Düren) Nucleospin RNA II Kit. The quality of total RNA was assessed with a 2100 Agilent Bioanalyzer (Agilent Technologies), and only samples of high quality (2100 Bioanalyzer RNA integrity value >7) were included in the analyses.

Array comparative genomic hybridization (arrayCGH)

An arrayCGH was carried out using an Agilent Human Genome CGH 244A Oligo Microarray. The samples were preprocessed according to an Agilent protocol (version 6.0, November 2008; Agilent Oligonucleotide Array-Based CGH for Genomic DNA Analysis). The gene copy-number data were processed as described earlier (19). Briefly, the data were LOESS normalized, segmented with circular binary segmentation using an R-package DNAcopy (20), and the regions with segment means more than two standard deviations from the mean (±0.67) of three self-versus-self control cell lines were called altered. The arrayCGH data are available under a Gene Expression Omnibus (GEO) accession number GSE108062. The significance of frequent alterations was assessed with the GISTIC v.2.0.21 software (the parameters used were q < 0.25, tamp = 20.67, tdel = 2−0.67, cap = 2, join segment size = 20). Copy-number polymorphic loci were excluded before data segmentation. The data were analyzed with the Anduril software framework (21). As due to the stringent criteria in arrayCGH analysis some previously reported alterations might be missed, the data were checked manually for consistency for selected regions with reported alterations.

Gene expression microarray analysis

Gene expression analysis was carried out on Illumina HT-12 human microarrays (Illumina Inc.). The RNA samples from the 45 cell lines were processed according to the manufacturer's recommendations. The gene expression data were quantile normalized using the lumi R-package (22). The gene expression data are available under a GEO accession number GSE108062. Downstream analysis of the gene expression data is described in detail below.

Custom panel design

A custom head and neck cancer panel was created using a Roche NimbleGen (Roche NimbleGen, Inc) Comprehensive Cancer Design consisting of 578 genes as a baseline. We customized this panel by analyzing these 578 genes in the cBioPortal (23, 24) for published mutations in head and neck cancers. In addition, information about the gene sequence alterations in head and neck cancers was searched from the PubMed. The genes with no known mutations in head and neck cancer were removed from the panel, and genes related to head and neck cancer were added, resulting in a head and neck cancer customized panel consisting of 694 genes (Supplementary Table S1). The gene list of the 694 genes was provided to Roche NimbleGen (Roche NimbleGen, Inc) for a SeqCap EZ Choice custom panel design. The exon locations were collated from ENSEMBL (v75), CCDS (29Nov2013), VEGA (v54), and RefSeq (13Aug2013). Overlaps were collapsed, and a single unified list of target exons was generated. These coordinates were further adjusted by expanding any targets less than 100 bp to a minimum length of 100 bp, centered in the middle of the exon. Any overlaps were then consolidated a second time. This coordinate file was used as the input for capture probe selection for a unique probe set containing unique probes as determined by the SSAHA (Sequence Search and Alignment by Hashing Algorithm). A probe was considered to match to the genome if there were five or fewer single-base insertions, deletions, or substitutions between the probe and the genome.

Next-generation sequencing

The genomic DNA was sheared with an S2-focused ultrasonicator (Covaris, Inc.). The library was prepared with a ThruPLEX DNA-seq Kit (Rubicon Genomics), and the targets were enriched using the custom SeqCap EZ Choice Library (Roche NimbleGen, Inc.). The samples were sequenced with an Illumina HiSeq sequencing system (Illumina Inc.) using a 100-nt paired ends read length. We reached an average coverage of 197 at the target regions.

The samples were aligned to hg19 using “bwa mem” (v. 0.7.10) with default parameters (25) and sorted with the Picard software (http://broadinstitute.github.io/picard). The targeted sequencing protocol greatly increases the likelihood of pseudoduplicated reads (i.e., reads that appear duplicated) and distinguishes real and false PCR duplicates. PCR duplicates were therefore not removed. Mutations were called using the VarScan v.2.3 software using parameters P = 0.95, somatic P = 0.05, strand-filter = 1, and min-var-frequency = 0.2 (26). Each cancer cell line was compared against the pooled sequences from the three control cell lines (normal skin fibroblasts from the same patient as UT_SCC_19A, 29 and 45). The germline variants included in the dbSNP and the 1000Genomes databases were removed after calling.

Drug-sensitivity testing, scoring, and hierarchical clustering

We used a compound library that consisted of 220 FDA-approved drugs as well as investigational compounds. A large proportion of the compounds were classified as signal transduction inhibitors (n = 94), mostly targeting either receptor or cytoplasmic kinases (Supplementary Table S2). Other classes that contained over 10 different compounds included alkylating agents, hormonal agents, antimetabolites, angiogenesis inhibitors, and topoisomerase inhibitor. Plerixafor and platinum salts including cisplatin and carboplatin were dissolved in water as dimethyl sulfoxide (DMSO) inactivates them while the rest of the compounds in the drug-sensitivity and resistance testing (DSRT) screening platform were dissolved in DMSO. The DSRT was carried out as described previously (27). Briefly, all compounds were tested over a 10,000-fold concentration range in five different concentrations in 384-well plates to generate quantitative and reliable dose-response data. Drugs were dispensed on plates by acoustic liquid handler (Echo 550, Labcyte) and dissolved in 5 μL of media. The cells were plated at a density of 1,000 cells/well in 20 μL volume by MultiDrop Combi peristaltic dispenser (Thermo Scientific) and incubated at 37°C and 5% CO2 for 72 hours. After 3 days cell viability was measured by CellTiter-Glo luminescent assay (Promega). DMSO and benzethonium chloride (100 μmol/L) were used as a negative and positive control, respectively.

The dose-response curves were modeled using a four-parameter logistic function, defined by the top and bottom asymptotes, the slope, and the half-maximal inhibition constant (IC50). In the curve fitting, the top asymptote of the curve was fixed to 100% viability, while the bottom asymptote was allowed to float between 0 and 75% (i.e., compounds causing less than 25% inhibition were considered inactive). To quantitatively profile the individual HNSCC cells in terms of their compound responses, we calculated the model-based drug-sensitivity score (DSS), which has been shown to improve the identification of cellular addictions and other druggable vulnerabilities in individual cancer patients over IC50 and other univariate response parameters (27, 28). The differential DSS (dDSS) was calculated by comparing the DSS of particular HNSCC cells to the mean DSS over all the cell lines. Unsupervised clustering of the DSS profiles across the tested compounds and HNSCC cell lines was performed using the Ward hierarchical complete-linkage clustering algorithm using Spearman and Manhattan distance measures for the drug and sample profiles, respectively.

Gene expression biomarker analysis for EGFR, MEK, and mTOR inhibitors

Drug classes were defined as follows: EGFR inhibitors consisted of afatinib, canertinib, gefitinib, and erlotinib; MEK inhibitors of refametinib, binimetinib, selumetinib, trametinib, pimasertib, and TAK-733; and mTOR inhibitors of OSI-027, AZD8055, sirolimus, temsirolimus, ridaforolimus, and everolimus. For each of these drug classes, we grouped the six cell lines with the highest class-specific DSS (median drug sensitivity calculated from the compound-specific DSSs in the drug class) into a high-sensitivity group, and the six cell lines with the lowest class-specific DSS into a low-sensitivity group. We compared the difference in the gene expression between the high and low-sensitivity groups, and the genes with a fold change of > 2 (group medians) were considered significantly differentially expressed (t test, P < 0.01). The P values for the significantly differentially expressed genes were adjusted by the Benjamini–Hochberg method (29) using “stats” R-package (R Foundation for Statistical Computing).

Somatic mutations as biomarkers for drug response

To assess the influence of a gene-specific mutation on drug sensitivity, the cell lines with a mutation were compared with the rest of the cell lines using the Wilcoxon rank-sum test (two-sided). The difference between two groups was assessed with permutation test using “coin” R-package (30). The P values were adjusted with the Benjamini–Hochberg (29) method as described above. Due to the functional relevance of stop-gain mutations to protein product, the identical analysis was performed to assess the influence of gene-specific stop-gain mutations on drug sensitivity. We restricted our analysis only to the most common mutations we observed in a gene panel of 694 genes.

Integrative analyses of drug sensitivity, gene expression, and gene copy numbers

The difference in the drug sensitivity between the cell lines with or without a copy-number alteration (CNA) was tested using the Wilcoxon rank-sum test, and the difference was considered significant at a false discovery rate (FDR) level q < 0.05 (Storey correction for multiple testing; ref. 31). The differences in drug sensitivity were quantified by subtracting the median DSS of the samples with a CNA from the median DSS of the samples without the CNA. The statistical testing was done for each drug separately. We further compared the gene expression data with the CNA data to find the genes whose expression is modified due to a CNA. By using the CNA data, we categorized the genes located in the same chromosome region into two groups, altered and nonaltered, and calculated the signal-to-noise ratio (32). The significance of the difference in gene expression between the two groups was assessed with a permutation test and corrected for multiple hypothesis using the Benjamini–Hochberg correction (29). Genes with an FDR q < 0.05 were considered significantly altered. Only CNAs that occurred in at least 10% of the cell lines were tested. Finally, the genes whose CNA status was significantly associated with both gene expression and drug sensitivity were further tested for an association between gene expression and drug-sensitivity profiles using the bootstrapCor function in the maigesPack R-package (−0.3 ≥ |r| ≥ 0.3; ref. 33) and adjusted with the Benjamini–Hochberg (29) method.

Real-time quantitative PCR (RT-qPCR)

mRNA was reverse transcribed to cDNA using an iScript cDNA synthesis kit (Bio-Rad) following the manufacturer's instructions. RT-qPCR analyses were done using the SYBR Green dye (LightCycler 480 DNA SYBR Green I Master, Cat. No. 04707516001, Roche Diagnostics GmbH). Quantitation was performed using LightCycler 480 384-multiwell plates and the LightCycler 480 instrument (Roche Diagnostics GmbH). The analyses were done in a triplicate for each gene. The primers used are listed in Supplementary Table S3. The data were analyzed with a Basic relative quantification program of the LightCycler 480 Software v.1.5 (Roche Diagnostics GbmH), based on the 2-ΔΔCT-method. A keratinocyte cell line, CCD 1106 KERTr (ATCC CRL-2309), served as the reference sample for gene expression normalization. GAPDH was selected as the reference gene for normalization.

siRNA transfection/drug-response measurement of cancer cell lines

To validate selected results from the drug screen, we silenced FAM83H (pool of siRNAs s50090, s50091, and s50092, Ambion) and RAB25 (pool of siRNAs SI03036544, SI02644089, SI02644082, and SI00126084, Qiagen) in UT-SCC-14 and UT-SCC-8 cell lines, respectively. A total of 12 nmol/L pooled siRNAs were used in three replicate experiments. Qiagen All Stars Negative (#1027281) served as a negative and Qiagen All Stars Hs Death Control (#1027299) as positive control. Cells were reverse transfected using Lipofectamine RNAiMAX (Thermo Fisher Scientific), and 24 hours later supplemented with nine different afatinib or erlotinib concentrations (0.1, 0.3, 1, 3.2, 10, 31.6, 100, 316.2, and 1,000 nmol/L or 1, 3.2, 10, 31.6, 100, 316.2, 1000, 3162.3, and 10,000 nmol/L, respectively). After 72 hours of drug treatment, the number of viable cells was measured using the CellTiter-Glo luminescent cell viability assay (Promega). Percent inhibition was calculated using the negative and positive controls using formula:

formula

To account for the siRNA effect on the cells, the percent inhibition value at the lowest concentration of each dose response was treated as the siRNA background, and this value was subtracted from values of the other concentrations. The percent inhibition data for each drug were then curve fitted with Levenberg–Marquardt method. The replicates were averaged for each concentration during the curve fitting. The IC50, slope, maximal and minimal response coming out of the curve fitting are then used to calculate the DSS (28).

HNSCC cell lines show selective responses to EGFR, MEK, and mTOR inhibitors

A high-throughput DSRT platform of 220 compounds showed selective responses across the 45 HNSCC cell lines (Supplementary Table S4) as measured with the DSS, a modified form of the area under the curve calculation that integrates multiple dose-response parameters for each drug. DSS therefore captures both the potency and the efficacy of the drug effect (Supplementary Table S4). Unsupervised hierarchical clustering of drug sensitivity over all 45 cell lines yielded eight clusters (Supplementary Fig. S1). Of these, group I was unresponsive to most of the 220 drugs tested. Three groups showed selectivity to EGFR and MEK inhibitors (groups II, VI, and VIII) as single agents, proposing synergistic effects for selected cell lines or in combination with an mTOR inhibitor (group VIII). Group V cell lines were responsive to mTOR inhibitors and mostly unresponsive to EGFR and MEK inhibitors.

The activity of EGFR tyrosine kinase inhibitors afatinib, canertinib, erlotinib, and gefitinib had a median DSS of >10, with canertinib (a pan-ErbB inhibitor) showing the highest activity (Supplementary Fig. S2; Supplementary Table S4). Many of the genomic alterations found in HNSCC may result in a persistent activation of the PI3K–AKT–mTOR pathway (11). In accordance with this, the cell lines often responded well to five of six tested mTOR inhibitors (AZD8055, everolimus, ridaforolimus, sirolimus, and temsirolimus) and to all tested PI3K/mTOR inhibitors (apitolisib, dactolisib, omipalisib, and PF-04691502). The median DSS to these drugs was >10 (Supplementary Figs. S1 and S2; Supplementary Table S4). Interestingly, all the other PI3K inhibitors except pictilisib did not show substantial efficacy (the median DSS <10). MEK inhibitors had variable efficacy, with a DSS >10 obtained with pimasertib, refametinib, TAK-733, and trametinib.

HNSCC cell lines resemble clinical HNSCC tumors in terms of genetic changes and display alterations that predict for drug response

The cell lines were subjected to a genome-wide analysis of gene copy-number gains and losses using an Agilent Human Genome CGH 244A Oligo Microarray and targeted to exome sequencing of 694 genes to correlate the genomic alterations with drug sensitivity. In general, the detected mutations, which occurred frequently in TP53, NOTCH1, FAT1, CSMD3, and CDKN2A, and the commonly observed CNAs (amplifications at 3q26, 7p11.2, 8q24, and 11q13 and deletions at 3p14, 8p23, 9p21, 9p23, and 18q), resemble those observed in clinical HNSCC (refs. 10–14, 34; Fig. 1; Supplementary Tables S5 and S6). In total, we detected 552 variants in 279 genes in the targeted HNSCC gene panel by next-generation sequencing (Supplementary Table S5). The number of variants among the studied 694 genes differed between the cell lines (range, 3–29; mean, 12). The UT-SCC-60A cell line had the largest and the UT-SCC-99 and UT-SCC-36 the lowest number of mutations. The number of mutations did not correlate with the primary tumor tumor–node–metastasis classification size category (P = 0.379; unpaired t test) or with cancer regional lymph node involvement (P = 0.887; unpaired t test).

Figure 1.

Genomic alterations in 45 HNSCC cell lines. A, Frequency plot of copy-number amplifications and deletions. Amplifications are shown in red and deletions in blue. The vertical axis shows the proportion of cell lines with a particular gain or loss. The horizontal black lines marks the 10% cutoffs that were used to include an alteration for further analyses. B, The most common mutations observed in the 45 HNSCC cell lines. The figure shows the genes that harbored ≥5 distinct mutations. The types of mutations are indicated with different shades of brown. The percentages (left) indicate the proportion of the cell lines with a mutation (one cell line may have ≥1 mutation). The proportion of HNSCCs with a mutation in the TCGA database is shown for comparison (the percentages on the right). Silent mutations were excluded. C, Coding mutations observed in NOTCH1 and FAT1 across the cell lines are shown. Splice-site mutations are excluded from the figure. Green indicates missense mutation, black truncating mutations, and violet other coding mutations. The image was created using MutationMapper at cBioPortal.

Figure 1.

Genomic alterations in 45 HNSCC cell lines. A, Frequency plot of copy-number amplifications and deletions. Amplifications are shown in red and deletions in blue. The vertical axis shows the proportion of cell lines with a particular gain or loss. The horizontal black lines marks the 10% cutoffs that were used to include an alteration for further analyses. B, The most common mutations observed in the 45 HNSCC cell lines. The figure shows the genes that harbored ≥5 distinct mutations. The types of mutations are indicated with different shades of brown. The percentages (left) indicate the proportion of the cell lines with a mutation (one cell line may have ≥1 mutation). The proportion of HNSCCs with a mutation in the TCGA database is shown for comparison (the percentages on the right). Silent mutations were excluded. C, Coding mutations observed in NOTCH1 and FAT1 across the cell lines are shown. Splice-site mutations are excluded from the figure. Green indicates missense mutation, black truncating mutations, and violet other coding mutations. The image was created using MutationMapper at cBioPortal.

Close modal

The genes that were mutated in at least five (≥10%) of the cell lines were compared with drug sensitivity (using the DSS) across the 45 cell lines. The results are shown in Supplementary Table S7. The analysis between CNA alterations and drug responses revealed 773 CNAs that were significantly associated with the drug sensitivity (Wilcoxon rank-sum test, Q < 0.1; Supplementary Table S8).

The influence of a CNA to gene expression was assessed by expression profiling using Illumina HT-12 microarrays. Of the 4,298 genes (1,952 deleted, 2,347 gained) that were located in the regions altered in at least 10% of the cell lines, 472 showed significant downregulation and 445 significant upregulation based on CNA cis effects at their loci (Supplementary Tables S9 and S10). The amplified and upregulated genes included PIK3CA at 3q26.3, EGFR at 7p11.2, CCND1, ORAOV1, PPFIA1, CTTN, and FADD at 11q13, and BIRC2, BIRC3, and YAP1 at 11q22. All of these genes are known to be located in frequently amplified chromosomal regions in HNSCC (10–13). The deleted and downregulated genes included CDKN2A at 9p21.3 and SMAD4 at the 18q21.2 region. A real-time quantitative PCR reaction of eight genes validated the gene expression microarray data (Supplementary Table S3).

Finally, to identify the genes that might be biomarkers for drug response, within the amplified and deleted regions, we integrated drug-sensitivity data with the gene expression data. Only those genes that showed marked CNAs in-cis and substantial changes in mRNA expression were included in this analysis. Significant drug-sensitivity-gene expression-CNA triads are presented in Supplementary Table S11. The most common genetic alterations and their association with drug response to EGFR, MEK and mTOR inhibitors for the 45 HNSCC cell lines are presented in Fig. 2.

Figure 2.

Associations between sensitivity to EGFR, MEK, and mTOR inhibitors and selected genetic alterations in HNSCC cell lines. The cell lines investigated are shown on the horizontal axis. On the vertical axis are shown a total number of mutations from a panel of 694 genes, as well as the investigated drugs and the genes. The top panel depicts the numbers and the types of mutations in each cell line. The center panel shows a heatmap of the differential DSSs (dDSS) for selected drugs. The bottom panel shows selected genomic alterations for frequently mutated or copy-number-altered genes. The ordering of the cell lines is based on the dDSS clustering.

Figure 2.

Associations between sensitivity to EGFR, MEK, and mTOR inhibitors and selected genetic alterations in HNSCC cell lines. The cell lines investigated are shown on the horizontal axis. On the vertical axis are shown a total number of mutations from a panel of 694 genes, as well as the investigated drugs and the genes. The top panel depicts the numbers and the types of mutations in each cell line. The center panel shows a heatmap of the differential DSSs (dDSS) for selected drugs. The bottom panel shows selected genomic alterations for frequently mutated or copy-number-altered genes. The ordering of the cell lines is based on the dDSS clustering.

Close modal

Systematic genomic analyses integrated with mTOR and MEK inhibitor response reveal candidate biomarkers

To find potential biomarkers for mTOR and MEK inhibitor sensitivity, we searched for somatic mutations and copy-number aberrations that associated with drug sensitivity from the analyses described above. In addition, the gene expression patterns of the six most sensitive and the six most resistant cell lines for these inhibitor classes were compared. These drugs were selected, because they showed high selectivity across all cell lines, and they are of clinical interest in the treatment of HNSCC.

A 24-gene signature discriminated the high and poorly responsive cell lines to six mTOR inhibitors (Fig. 3A; Supplementary Table S12), from which everolimus and temsirolimus are currently being evaluated as therapeutic options in combination with chemotherapy, radiotherapy, or other molecularly targeted drugs in HNSCC (35). One of these genes was oxysterol binding protein like 1A (OSBPL1A), a gene involved in lipid metabolism. The integrative analysis of CNA-gene expression and DSS revealed that the expression of OSBPL1A was affected by deletion in a group of poorly responsive cell lines. In comparison with the gene expression profiles of the six most sensitive and resistant cell lines for six MEK inhibitors, 11 genes, including EFNB1 and STAT6, showed higher levels of expression in the cell lines sensitive to MEK inhibitors as compared with cell lines resistant to these drugs (Fig. 3B; Supplementary Table S12).

Figure 3.

Selected genetic alterations that are associated with sensitivity to mTOR and MEK inhibitors. A and B, Unsupervised clustering of differentially expressed genes associated with sensitivity to mTOR inhibitors (OSI-027, AZD8055, sirolimus, temsirolimus, ridaforolimus, and everolimus) and MEK inhibitors (refametinib, binimetinib, selumetinib, trametinib, pimasertib, and TAK-733), respectively. Hierarchical clustering is based on the Euclidian distance calculated with an average linkage clustering algorithm. The color coding was scaled separately for each row to demonstrate the differences in gene expression. Red color indicates overexpression and green underexpression. The high and the low drug-sensitivity groups each contain six cell lines with the highest and the lowest class-specific DSS. C, An oncoprint of NOTCH1, NAV3, and PTPRD alterations across the 45 HNSCC cell lines. Individual genes are represented as rows, and the cell lines are represented as columns. The cell line order is based on sensitivity to refametinib. The oncoprint was created by using the cBioPortal. D, Sensitivity to temsirolimus, trametinib, and refametinib expressed as DSS value of cell lines, grouped according to FAT1, CDKN2A, and NOTCH1 mutation status. Wilcoxon rank-sum test P = 0.02 (FAT1), 0.04 (CDKN2A), and 0.03 (NOTCH1).

Figure 3.

Selected genetic alterations that are associated with sensitivity to mTOR and MEK inhibitors. A and B, Unsupervised clustering of differentially expressed genes associated with sensitivity to mTOR inhibitors (OSI-027, AZD8055, sirolimus, temsirolimus, ridaforolimus, and everolimus) and MEK inhibitors (refametinib, binimetinib, selumetinib, trametinib, pimasertib, and TAK-733), respectively. Hierarchical clustering is based on the Euclidian distance calculated with an average linkage clustering algorithm. The color coding was scaled separately for each row to demonstrate the differences in gene expression. Red color indicates overexpression and green underexpression. The high and the low drug-sensitivity groups each contain six cell lines with the highest and the lowest class-specific DSS. C, An oncoprint of NOTCH1, NAV3, and PTPRD alterations across the 45 HNSCC cell lines. Individual genes are represented as rows, and the cell lines are represented as columns. The cell line order is based on sensitivity to refametinib. The oncoprint was created by using the cBioPortal. D, Sensitivity to temsirolimus, trametinib, and refametinib expressed as DSS value of cell lines, grouped according to FAT1, CDKN2A, and NOTCH1 mutation status. Wilcoxon rank-sum test P = 0.02 (FAT1), 0.04 (CDKN2A), and 0.03 (NOTCH1).

Close modal

Regarding CNAs, deletion of PTPRD at 9p24.1, present in nine cell lines, was statistically significantly associated with poor sensitivity to MEK inhibitor refametinib as assessed by Wilcoxon rank-sum test (Q < 0.1; Supplementary Table S8; Fig. 3C).

We found no significant associations between responses to mTOR or MEK inhibitors with mutations after correcting for multiple hypothesis testing (permutation test Q > 0.1). However, as specific genes have mutations in relatively small number of cell lines in our cohort, a larger series of samples is needed to further assess the statistical significance of the alterations. As an example, UT-SCC cell lines that harbored a stop-gain mutation in FAT1 (present in 9% of the cell lines) showed a tendency for a higher sensitivity to temsirolimus as compared with the other cell lines (Wilcoxon rank-sum test P < 0.05, Fig. 3D; Supplementary Table S7).

Mutations in NOTCH1, which is one of the most commonly mutated genes in HNSCC, were associated with a poor response to refametinib (Wilcoxon rank-sum test P < 0.05; Fig. 3C and D). Interestingly, mutations in NAV3 were associated with poorer efficacy of four of the six MEK inhibitors tested (Wilcoxon rank-sum test P < 0.05; Supplementary Table S7; Fig. 3C). Cell lines with CDKN2A mutation, which occurred in 20% of our cell lines, had tendency for poor sensitivity to the MEK inhibitor trametinib (Wilcoxon rank-sum test P < 0.05, Fig. 3D; Supplementary Table S7). On the other hand, CDKN2A deletions or decreased expression did not show significant associations with drug response.

Novel biomarkers predict response to EGFR inhibitors

EGFR status has not been considered as a reliable biomarker for response to EGFR inhibitors in HNSCC (36). In our study, cell lines with EGFR amplification and overexpression were weakly associated with sensitivity to selected inhibitors, such as erlotinib (r = 0.36) and canertinib (r = 0.30; Supplementary Table S11). Most cell lines with increased EGFR copy number showed sensitivity to EGFR inhibitors, but in line with previous studies, the result was not conclusive (Fig. 4A).

Figure 4.

Selected genetic alterations associated with response to EGFR inhibitors. A, A waterfall plot showing the cell line median sensitivity to EGFR inhibitors across the 45 HNSCC cell lines. The cell lines that harbor a CNA gain for EGFR are shaded. Cell lines have been ordered by the differential DSS (dDSS) calculated by comparing the DSS of particular HNSCC cells to the mean DSS over all the cell lines. B, Unsupervised clustering of differentially expressed genes associated with sensitivity to EGFR inhibitors (afatinib, canertinib, gefitinib, and erlotinib). Hierarchical clustering is based on a Euclidian distance obtained with an average linkage clustering algorithm. The color coding was scaled separately for each row to better show differences in gene expression. Red color indicates overexpression and green underexpression. The high and the low drug-sensitivity groups each contain six cell lines with the highest and the lowest DSS to EGFR inhibitors. C, Box plots showing expression of selected genes involved with cell membrane trafficking. Expression of each gene is associated with sensitivity to EGFR inhibitors. D, An oncoprint of NOTCH1, NAV3, MYC, and FAM83H alterations across the 45 HNSCC cell lines. Individual genes are represented as rows, and cell lines as columns. The cell line order is based on afatinib sensitivity. The oncoprint was created using the cBioPortal. E, Box plots showing expression of selected three genes in the CCLE data set related to erlotinib sensitivity. High expression of each gene (RAB25, TMEM125, and FAM83H) was significantly associated with sensitivity to erlotinib (Wilcoxon rank-sum test; ns, not significant; *, P = 0.003; **, P < 0.001).

Figure 4.

Selected genetic alterations associated with response to EGFR inhibitors. A, A waterfall plot showing the cell line median sensitivity to EGFR inhibitors across the 45 HNSCC cell lines. The cell lines that harbor a CNA gain for EGFR are shaded. Cell lines have been ordered by the differential DSS (dDSS) calculated by comparing the DSS of particular HNSCC cells to the mean DSS over all the cell lines. B, Unsupervised clustering of differentially expressed genes associated with sensitivity to EGFR inhibitors (afatinib, canertinib, gefitinib, and erlotinib). Hierarchical clustering is based on a Euclidian distance obtained with an average linkage clustering algorithm. The color coding was scaled separately for each row to better show differences in gene expression. Red color indicates overexpression and green underexpression. The high and the low drug-sensitivity groups each contain six cell lines with the highest and the lowest DSS to EGFR inhibitors. C, Box plots showing expression of selected genes involved with cell membrane trafficking. Expression of each gene is associated with sensitivity to EGFR inhibitors. D, An oncoprint of NOTCH1, NAV3, MYC, and FAM83H alterations across the 45 HNSCC cell lines. Individual genes are represented as rows, and cell lines as columns. The cell line order is based on afatinib sensitivity. The oncoprint was created using the cBioPortal. E, Box plots showing expression of selected three genes in the CCLE data set related to erlotinib sensitivity. High expression of each gene (RAB25, TMEM125, and FAM83H) was significantly associated with sensitivity to erlotinib (Wilcoxon rank-sum test; ns, not significant; *, P = 0.003; **, P < 0.001).

Close modal

Interestingly, when we compared the gene expression between cell lines responsive and resistant to EGFR inhibitors, the cell lines sensitive to EGFR inhibitors showed high mRNA levels for several genes associated with membrane trafficking and transport, such as RAB25, CLIC3, SYK, SLC31A2, LYPD3, TMEM125, and TSPAN1 (Fig. 4B and C). Knockdown of RAB25 slightly reduced the sensitivity of UT-SCC-8 cells to erlotinib and afatinib (Supplementary Fig. S3A–S3D and S3I).

In the integrative analysis of drug sensitivity, gene expression, and gene copy numbers, FAM83H overexpression, which was caused by gene amplification, correlated with enhanced sensitivity to EGFR inhibitors afatinib (r = 0.53, permutation P = 0.0001, Q = 0.2, Fig. 4D), erlotinib (r = 0.53), canertinib (r = 0.51), and gefitinib (r = 0.49; permutation P < 0.001, Q > 0.2; Supplementary Table S11). This gene is amplified in 12% of HNSCCs (13). Moreover, FAM83H silencing made UT-SCC-14 cells more resistant to afatinib and erlotinib (Supplementary Fig. S3E–S3I).

To validate the gene expression results for RAB25, TMEM125, and FAM83H in an independent data set, we determined drug-sensitivity calls (sensitive, intermediate, or resistant) in the Cancer Cell Line Encyclopedia (CCLE; ref. 37), which provides genetic and expression data for 490 cell lines together with their response to EGFR inhibitor erlotinib. We used 3-bin k-means clustering of the −logIC50 values. The sensitivity to erlotinib and expression of RAB25, TMEM125, and FAM83H were significantly associated in the CCLE data set (Fig. 4E).

Similar to MEK inhibitors, mutations in NAV3 and NOTCH1 showed tendency with a poor response to EGFR inhibitors (P < 0.05; Supplementary Table S7; Fig. 4D) discriminating a group of cell lines with poorer responses to both EGFR and MEK inhibitors. In addition, several CNA alterations were associated with response to EGFR inhibitors afatinib (Fig. 4D), canertinib, gefitinib, and erlotinib (Supplementary Table S8). Cell lines with MYC amplification showed tendency for better response to EGFR inhibitors gefitinib, afatinib, and erlotinib as compared with the other cell lines (Supplementary Table S8). FAM83H was coamplified with MYC (Fig. 4D), which might explain the association between MYC amplification and EGFR inhibitor response. In line with these results, there was also a significant correlation between FAM83H and MYC amplification in The Cancer Genome Atlas (TCGA) HNSCC study (13) when we analyzed the cBioPortal database (Fisher exact test, P < 0.001).

Apart from cetuximab and immune checkpoint inhibitors (6, 7, 38), no other targeted drugs to treat advanced HNSCC are currently available. We studied 45 HNSCC cell lines established from HPV-negative HNSCCs on a high-throughput drug-testing platform (DSRT) and combined the drug-sensitivity information with detailed genomic and transcriptomic data, which allowed us to find novel molecular associations for drug responses. We found several genomic alterations such as mutations in NOTCH1 and FAT1 and deletions in PTPRD1, which showed tendency with poor efficacy of EGFR and MEK inhibitors. In addition, overexpression of FAM83H and amplification of MYC showed tendency for improved sensitivity to EGFR inhibitors. To our knowledge, the present study is the largest study on HNSCC that integrates genetic information with drug responses.

A limitation of this study is that only cell lines were tested, and the number of single alterations was low, affecting the power of the statistical analyses. However, testing of a large number of compounds and biomarkers is challenging in clinical trials or in xenograft models due to financial and other constraints. Repeated expansion of the cell lines likely results in adaptive changes in gene expression, but many of the cell lines used in this study had low passage numbers. Prior studies on cancer cell lines generally agree with their value in drug discovery for molecularly targeted therapies (37, 39–43). Genomic characterization of the cell lines corresponded well with the mutation spectrum observed in human HNSCCs (13), suggesting that the cell lines recapitulate oncogenic alterations observed in clinical tumor samples. Recognizing that the translation of potential associations detected in preclinical models to clinically useful biomarkers may be challenging and requires validation (42, 43), the integration of data obtained from DSRT and genomic characterization appears a powerful strategy to identify systemic treatment options and potential biomarkers (27, 28).

EGFR inhibitors are currently the only targeted kinase inhibitors that have been approved for the treatment of advanced HNSCC and have modest efficacy. In HNSCC, EGFR expression or an increased EGFR gene copy number has been extensively studied as biomarkers for response to EGFR inhibitors, but they are not predictive or prognostic (36). In our study, EGFR amplification or overexpression was weakly associated with sensitivity to selected EGFR inhibitors, but in line with the previous studies, the results were not conclusive. On the other hand, cell lines sensitive for EGFR inhibitors had high mRNA levels for genes involved in membrane trafficking and receptor and membrane transport, suggesting a role for the plasma membrane protein organization and EGFR trafficking in drug response. For example, the EGFR inhibitor–sensitive cell lines showed 8 times higher RAB25 mRNA levels as compared with the insensitive cell lines. In agreement with this finding, Rab25 was found to influence EGFR endocytosis and gefitinib efficacy (44). Interestingly, the cell lines with MYC amplification responded significantly better to EGFR inhibitors as compared with other cell lines. In a series with non–small cell lung cancer patients, a high MYC copy number correlated with high sensitivity to EGFR inhibitors (45).

An example of a novel molecular association with EGFR inhibitor response in the present study is amplification and mRNA overexpression of FAM83H. The FAM83 proteins were recently identified as novel transforming oncogenes that promote resistance to EGFR tyrosine kinase inhibitors in breast cancer (46). However, suggesting that the effects of FAM83 proteins on drug sensitivity might vary for different FAM83 family members and might be tissue-type dependent, the detailed molecular mechanisms that explain these associations remain to be elucidated.

Head and neck cancers harbor a wide spectrum of genetic alterations, and observations from several studies point to the importance of mutations in TP53, NOTCH1, CDKN2A, PIK3CA, FBXW7, and HRAS in the molecular pathogenesis of HNSCC (10–13). In the present study, NOTCH1 was the second most commonly mutated gene (present in 29% of the cell lines), and the mutations were mostly found in cell lines with poor sensitivity to EGFR and MEK inhibitors. NOTCH1 is regulated by TP53, and it has both oncogenic and tumorigenic functions (47–49). NOTCH1 is involved in squamous cell differentiation, and in keratinocytes increased NOTCH activity reduces proliferation and promotes differentiation, whereas loss of NOTCH1 promotes carcinogenesis (47–49). NOTCH1 activity and concurrent downstream activation of MYC are associated with resistance of breast cancer cells to PI3K inhibitors (50). Only 3 of the 24 cell lines harboring mutations either in NOTCH1 or in the PI3K/mTOR pathway had mutations in both, which is in line with previous clinical data showing no correlation between the PI3K/mTOR pathway and NOTCH1 mutations (11). Taken together, the results support an important role for NOTCH1 signaling in the genesis of some HNSCC and as a potential drug target and biomarker.

In summary, the current comprehensive genomic analysis integrated with high-throughput drug testing on a large panel of HNSCC cell lines identified several biomarkers for EGFR, MEK, and mTOR inhibitor efficacy in HNSCC. These biomarkers included genetic alterations such as NOTCH1 and NAV3 mutations, MYC and FAM83H amplifications, and PTPRD deletion. These potential biomarkers deserve further validation in clinical series, because robust biomarkers for EGFR, MEK, and mTOR inhibitor efficacy are lacking.

H. Joensuu is Vice President at and is a consultant/advisory board member for Orion Pharma and has ownership interest (including stock, patents, etc.) in Sartar Therapeutics. No potential conflicts of interest were disclosed by the other authors.

Conception and design: T. Lepikhova, P.-R. Karhemo, H. Joensuu, Outi Monni

Development of methodology: T. Lepikhova, R. Grénman, K. Wennerberg

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): T. Lepikhova, P.-R. Karhemo, A. Murumägi, E. Kulesskiy, H. Sihto, R. Grénman, S.M. Syrjänen, O. Kallioniemi, H. Joensuu, Outi Monni

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): T. Lepikhova, P.-R. Karhemo, R. Louhimo, B. Yadav, E. Kulesskiy, M. Kivento, H. Sihto, T. Aittokallio, K. Wennerberg, H. Joensuu, Outi Monni

Writing, review, and/or revision of the manuscript: T. Lepikhova, P.-R. Karhemo, R. Louhimo, B. Yadav, A. Murumägi, E. Kulesskiy, M. Kivento, H. Sihto, R. Grénman, S.M. Syrjänen, O. Kallioniemi, T. Aittokallio, K. Wennerberg, H. Joensuu, Outi Monni

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): P.-R. Karhemo, M. Kivento, O. Kallioniemi, H. Joensuu, Outi Monni

Study supervision: H. Joensuu, Outi Monni

Other (coordination and detailed planning of the project): T. Lepikhova

Other (data integration): B. Yadav

Other (HPV genotyping): S.M. Syrjänen

We thank the Biomedicum Functional Genomics Unit (FuGU), FIMM High-Throughput Biomedicine Unit, and FIMM Sequencing Unit at the University of Helsinki, Finland, for excellent technical assistance.

This work was financially supported by Academy of Finland (O. Monni, H. Joensuu, T. Aittokallio, and K. Wennerberg; Center of Excellence for Translational Cancer Biology, O. Kallioniemi, H. Joensuu), Jane & Aatos Erkko Foundation (O. Monni, H. Joensuu, and K. Wennerberg), Finnish Cancer Society (O. Monni, H. Joensuu, T. Aittokallio, K. Wennerberg, and O. Kallioniemi), Sigrid Juselius Foundation (H. Joensuu, K. Wennerberg, and O. Kallioniemi), and Research Funds of Helsinki University Hospital (H. Joensuu).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ferlay
J
,
Soerjomataram
I
,
Dikshit
R
,
Eser
S
,
Mathers
C
,
Rebelo
M
, et al
Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012
.
Int J Cancer
2015
;
136
:
E359
86
.
2.
Fury
MG
,
Pfister
DG
. 
Current recommendations for systemic therapy of recurrent and/or metastatic head and neck squamous cell cancer
.
J Natl Compr Canc Netw
2011
;
9
:
681
9
.
3.
Belcher
R
,
Hayes
K
,
Fedewa
S
,
Chen
AY
. 
Current treatment of head and neck squamous cell cancer
.
J Surg Oncol
2014
;
110
:
551
74
.
4.
Dasari
S
,
Tchounwou
PB
. 
Cisplatin in cancer therapy: molecular mechanisms of action
.
Eur J Pharmacol
2014
;
740
:
364
78
.
5.
Moy
JD
,
Moskovitz
JM
,
Ferris
RL
. 
Biological mechanisms of immune escape and implications for immunotherapy in head and neck squamous cell carcinoma
.
Eur J Cancer
2017
;
76
:
152
66
.
6.
Bonner
JA
,
Harari
PM
,
Giralt
J
,
Azarnia
N
,
Shin
DM
,
Cohen
RB
, et al
Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck
.
N Engl J Med
2006
;
354
:
567
78
.
7.
Cohen
EE
. 
Role of epidermal growth factor receptor pathway-targeted therapy in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck
.
J Clin Oncol
2006
;
24
:
2659
65
.
8.
Leemans
CR
,
Braakhuis
BJ
,
Brakenhoff
RH
. 
The molecular biology of head and neck cancer
.
Nat Rev Cancer
2011
;
11
:
9
22
.
9.
Agrawal
N
,
Frederick
MJ
,
Pickering
CR
,
Bettegowda
C
,
Chang
K
,
Li
RJ
, et al
Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1
.
Science
2011
;
333
:
1154
7
.
10.
Pickering
CR
,
Zhang
J
,
Yoo
SY
,
Bengtsson
L
,
Moorthy
S
,
Neskey
DM
, et al
Integrative genomic characterization of oral squamous cell carcinoma identifies frequent somatic drivers
.
Cancer Discov
2013
;
3
:
770
81
.
11.
Lui
VW
,
Hedberg
ML
,
Li
H
,
Vangara
BS
,
Pendleton
K
,
Zeng
Y
, et al
Frequent mutation of the PI3K pathway in head and neck cancer defines predictive biomarkers
.
Cancer Discov
2013
;
3
:
761
9
.
12.
Seiwert
TY
,
Zuo
Z
,
Keck
MK
,
Khattri
A
,
Pedamallu
CS
,
Stricker
T
, et al
Integrative and comparative genomic analysis of HPV-positive and HPV-negative head and neck squamous cell carcinomas
.
Clin Cancer Res
2015
;
21
:
632
41
.
13.
Cancer Genome Atlas Network
. 
Comprehensive genomic characterization of head and neck squamous cell carcinomas
.
Nature
2015
;
517
:
576
82
.
14.
McDermott
U
,
Downing
JR
,
Stratton
MR
. 
Genomics and the continuum of cancer care
.
N Engl J Med
2011
;
364
:
340
50
.
15.
Ang
KK
,
Harris
J
,
Wheeler
R
,
Weber
R
,
Rosenthal
DI
,
Nguyen-Tan
PF
, et al
Human papillomavirus and survival of patients with oropharyngeal cancer
.
N Engl J Med
2010
;
363
:
24
35
.
16.
Morris
LG
,
Chandramohan
R
,
West
L
,
Zehir
A
,
Chakravarty
D
,
Pfister
DG
, et al
The molecular landscape of recurrent and metastatic head and neck cancers: insights from a precision oncology sequencing platform
.
JAMA Oncol
2017
;
3
:
244
55
.
17.
Grenman
R
,
Pekkola-Heino
K
,
Joensuu
H
,
Aitasalo
K
,
Klemi
P
,
Lakkala
T
. 
UT-MUC-1, a new mucoepidermoid carcinoma cell line, and its radiosensitivity
.
Arch Otolaryngol Head Neck Surg
1992
;
118
:
542
7
.
18.
Lansford
C
,
Grenman
R
,
Bier
H
,
Somers
KD
,
Kim
SY
,
Whiteside
TL
, et al
Head and Neck Cancers
.
In
:
Masters
JaPB
.,
editor
.
Human cell culture. Vol II: Cancer cell lines Part 2
.
Norwell, MA
:
Kluwer Academic Publishers
; 
1999
.
pp.
185
256
.
19.
Louhimo
R
,
Lepikhova
T
,
Monni
O
,
Hautaniemi
S
. 
Comparative analysis of algorithms for integration of copy number and expression data
.
Nat Methods
2012
;
9
:
351
5
.
20.
Olshen
AB
,
Venkatraman
ES
,
Lucito
R
,
Wigler
M
. 
Circular binary segmentation for the analysis of array-based DNA copy number data
.
Biostatistics
2004
;
5
:
557
72
.
21.
Ovaska
K
,
Laakso
M
,
Haapa-Paananen
S
,
Louhimo
R
,
Chen
P
,
Aittomaki
V
, et al
Large-scale data integration framework provides a comprehensive view on glioblastoma multiforme
.
Genome Med
2010
;
2
:
65
.
22.
Lin
SM
,
Du
P
,
Huber
W
,
Kibbe
WA
. 
Model-based variance-stabilizing transformation for Illumina microarray data
.
Nucleic Acids Res
2008
;
36
:
e11
.
23.
Cerami
E
,
Gao
J
,
Dogrusoz
U
,
Gross
BE
,
Sumer
SO
,
Aksoy
BA
, et al
The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data
.
Cancer Discov
2012
;
2
:
401
4
.
24.
Gao
J
,
Aksoy
BA
,
Dogrusoz
U
,
Dresdner
G
,
Gross
B
,
Sumer
SO
, et al
Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal
.
Sci Signal
2013
;
6
:
pl1
.
25.
Li
H
,
Durbin
R
. 
Fast and accurate short read alignment with Burrows-Wheeler transform
.
Bioinformatics
2009
;
25
:
1754
60
.
26.
Koboldt
DC
,
Zhang
Q
,
Larson
DE
,
Shen
D
,
McLellan
MD
,
Lin
L
, et al
VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing
.
Genome Res
2012
;
22
:
568
76
.
27.
Pemovska
T
,
Kontro
M
,
Yadav
B
,
Edgren
H
,
Eldfors
S
,
Szwajda
A
, et al
Individualized systems medicine strategy to tailor treatments for patients with chemorefractory acute myeloid leukemia
.
Cancer Discov
2013
;
3
:
1416
29
.
28.
Yadav
B
,
Pemovska
T
,
Szwajda
A
,
Kulesskiy
E
,
Kontro
M
,
Karjalainen
R
, et al
Quantitative scoring of differential drug sensitivity for individually optimized anticancer therapies
.
Sci Rep
2014
;
4
:
5193
.
29.
Benjamini
Y
,
Hochberg
Y
. 
Controlling the false discovery rate: a practical and powerful approach to multiple testing
.
J R Statist Soc B
1995
;
57
:
289
300
.
30.
Hothorn
T
,
Hornik
K
,
van de Wiel
M
,
Zeileis
A
. 
Implementing a class of permutation tests: the coin package
.
J Stat Softw
2008
;
28
:
1
23
.
31.
Storey
JD
,
Taylor
JE
,
Siegmund
D
. 
Strong control, conservative point estimation and simultaneous conservative consistency of false discovery rates: a unified approach
.
J R Statist Soc B
2004
;
66
:
187
205
.
32.
Hautaniemi
S
,
Ringnér
M
,
Kauraniemi
P
,
Autio
R
,
Edgren
H
,
Yli-Harja
O
, et al
A strategy for identifying putative causes of gene expression variation in human cancers
.
J Franklin Inst
2004
;
341
:
77
88
.
33.
Heyer
LJ
,
Kruglyak
S
,
Yooseph
S
. 
Exploring expression data: identification and analysis of coexpressed genes
.
Genome Res
1999
;
9
:
1106
15
.
34.
Jarvinen
AK
,
Autio
R
,
Haapa-Paananen
S
,
Wolf
M
,
Saarela
M
,
Grenman
R
, et al
Identification of target genes in laryngeal squamous cell carcinoma by high-resolution copy number and gene expression microarray analyses
.
Oncogene
2006
;
25
:
6997
7008
.
35.
Azoury
SC
,
Gilmore
RC
,
Shukla
V
. 
Molecularly targeted agents and immunotherapy for the treatment of head and neck squamous cell cancer (HNSCC)
.
Discov Med
2016
;
21
:
507
16
.
36.
Bossi
P
,
Resteghini
C
,
Paielli
N
,
Licitra
L
,
Pilotti
S
,
Perrone
F
. 
Prognostic and predictive value of EGFR in head and neck squamous cell carcinoma
.
Oncotarget
2016
;
7
:
74362
79
.
37.
Barretina
J
,
Caponigro
G
,
Stransky
N
,
Venkatesan
K
,
Margolin
AA
,
Kim
S
, et al
The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity
.
Nature
2012
;
483
:
603
7
.
38.
Kim
HS
,
Lee
JY
,
Lim
SH
,
Park
K
,
Sun
JM
,
Ko
YH
, et al
Association between PD-L1 and HPV status and the prognostic value of PD-L1 in oropharyngeal squamous cell carcinoma
.
Cancer Res Treat
2016
;
48
:
527
36
.
39.
Sharma
SV
,
Haber
DA
,
Settleman
J
. 
Cell line-based platforms to evaluate the therapeutic efficacy of candidate anticancer agents
.
Nat Rev Cancer
2010
;
10
:
241
53
.
40.
Solit
DB
,
Garraway
LA
,
Pratilas
CA
,
Sawai
A
,
Getz
G
,
Basso
A
, et al
BRAF mutation predicts sensitivity to MEK inhibition
.
Nature
2006
;
439
:
358
62
.
41.
Garnett
MJ
,
Edelman
EJ
,
Heidorn
SJ
,
Greenman
CD
,
Dastur
A
,
Lau
KW
, et al
Systematic identification of genomic markers of drug sensitivity in cancer cells
.
Nature
2012
;
483
:
570
5
.
42.
Hammerman
PS
,
Hayes
DN
,
Grandis
JR
. 
Therapeutic insights from genomic studies of head and neck squamous cell carcinomas
.
Cancer Discov
2015
;
5
:
239
44
.
43.
Kang
H
,
Kiess
A
,
Chung
CH
. 
Emerging biomarkers in head and neck cancer in the era of genomics
.
Nat Rev Clin Oncol
2015
;
12
:
11
26
.
44.
Jo
U
,
Park
KH
,
Whang
YM
,
Sung
JS
,
Won
NH
,
Park
JK
, et al
EGFR endocytosis is a novel therapeutic target in lung cancer with wild-type EGFR
.
Oncotarget
2014
;
5
:
1265
78
.
45.
Cappuzzo
F
,
Varella-Garcia
M
,
Rossi
E
,
Gajapathy
S
,
Valente
M
,
Drabkin
H
, et al
MYC and EIF3H Coamplification significantly improve response and survival of non-small cell lung cancer patients (NSCLC) treated with gefitinib
.
J Thorac Oncol
2009
;
4
:
472
8
.
46.
Lee
SY
,
Meier
R
,
Furuta
S
,
Lenburg
ME
,
Kenny
PA
,
Xu
R
, et al
FAM83A confers EGFR-TKI resistance in breast cancer cells and in mice
.
J Clin Invest
2012
;
122
:
3211
20
.
47.
Andersson
ER
,
Sandberg
R
,
Lendahl
U
. 
Notch signaling: simplicity in design, versatility in function
.
Development
2011
;
138
:
3593
612
.
48.
Dotto
GP
. 
Notch tumor suppressor function
.
Oncogene
2008
;
27
:
5115
23
.
49.
Nicolas
M
,
Wolfer
A
,
Raj
K
,
Kummer
JA
,
Mill
P
,
van Noort
M
, et al
Notch1 functions as a tumor suppressor in mouse skin
.
Nat Genet
2003
;
33
:
416
21
.
50.
Muellner
MK
,
Uras
IZ
,
Gapp
BV
,
Kerzendorfer
C
,
Smida
M
,
Lechtermann
H
, et al
A chemical-genetic screen reveals a mechanism of resistance to PI3K inhibitors in cancer
.
Nat Chem Biol
2011
;
7
:
787
93
.