Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Sep 1.
Published in final edited form as: Head Neck. 2010 Sep;32(9):1143–1160. doi: 10.1002/hed.21311

Integrative molecular characterization of head and neck cancer cell model genomes

Ivy FL Tsui 1,2, Cathie Garnis 1,3
PMCID: PMC2930055  NIHMSID: NIHMS157427  PMID: 20014447

Abstract

Background

Cell lines are invaluable model systems for the investigation of cancer. Knowledge of the molecular alterations that exist within cell models is required to define the mechanisms governing cellular phenotypes.

Methods

Five tongue squamous cell carcinomas cell lines and one submaxillary salivary gland epidermoid carcinoma cell line were analyzed for copy number and mRNA expression by tiling-path DNA microarrays and Agilent Whole Human Genome Oligoarrays, respectively.

Results

Integrative analysis of genetic and expression alterations revealed the molecular landscape of each cell line. Molecular results for individual cell lines and across all samples have been summarized and made available for easy reference.

Conclusion

Our integrative genomic analyses have defined the DNA and RNA alterations for each individual line. These data will be useful to anyone modelling oral cancer behaviour, providing a molecular context that will be useful for deciphering cell phenotypes.

Keywords: oral cancer, head and neck cancer, cell models, copy number alteration, RNA expression alteration

INTRODUCTION

Head and neck squamous cell carcinoma (HNSCC) is the eighth most common cancer in the world.(1) It is a heterogeneous disease primarily affecting the oral cavity, salivary glands, oropharynx, hypopharynx, and larynx. Model systems, such as cell lines are often used as research tools to study many cancer types, including HNSCC.(2, 3) They allow for manipulation of tumour cells in a laboratory setting. However, tumourigenic cell lines in such studies have often not been fully characterized at the molecular level. Like tumours themselves, cancer cell lines can vary greatly with respect to their genetic background. Analysis of cell phenotypes without concurrent analysis of underlying molecular changes represents a lost opportunity to understand the mechanistic basis for disease.

One characteristic of most solid tumours is genomic instability. This is demonstrated by the numerous DNA gains and deletions present in tumour genomes.(4) DNA alterations can lead to aberrant expression of oncogenes and tumour suppressor genes that drive tumourigenesis. For HNSCC development, gene alterations such as loss of chromosomes 3p and 9p, mutation of p53, loss of 13q, and high-level amplification of CCND1 have been associated with premalignant disease stages.(58) Loss of chromosomes 4q and 8p, on the other hand, are believed to occur as later events in tumourigenesis.(9) Such DNA alterations, including high-level amplification of CCND1,(10) often leads to overexpression, while homozygous deletion of CDKN2A leads to silencing of p16INK4A in oral tumours.(11)

Established HNSCC cell lines — particularly SCC-4, SCC-9, SCC-15, SCC-25, A253, and Cal27 — are widely used as models of head and neck cancer. A preliminary search in PubMed reveals 407 publications that use at least one of these lines, demonstrating their importance as tools for modeling biochemical, immunological, and pharmacological behaviours. These various behaviours could be influenced by the molecular alterations that exist in each line, yet few efforts have been made to characterize such changes. Previously, genetic alterations of four of the OSCC cell lines (SCC-4, SCC-9, SCC-15, and SCC-25) have been characterized by cDNA microarrays with low resolution and with no data on intragenic regions.(12) Thus, comprehensive characterization of genomic alterations in the most commonly used cell models are needed. In addition, a priori knowledge of both genomic and gene expression changes for a given cell line would facilitate selection of the most appropriate model system for a given in vitro analysis. In this report we provide full characterization of genome and transcriptome alterations in six commonly used HNSCC cell lines. These data will serve as an excellent resource when designing future experiments that attempt to model HNSCC behaviour.

MATERIALS AND METHODS

Cell lines

Five tongue squamous cell carcinomas cell lines (SCC-4,(13) SCC-9,(13) SCC-25,(13) SCC-15,(13) and Cal27(14)) and one submaxillary salivary gland epidermoid carcinoma cell line (A-253)(15) were purchased from the American Type Culture Collection (ATCC). All cell lines were cultured according to ATCC recommendations. Cells were harvested for DNA and RNA extractions when they reached ~ 80% confluency. Genomic DNA was extracted by the standard phenol-chloroform extraction protocol, and total RNA was extracted using the TRIzol (Invitrogen, Carlsbad, CA) protocol followed by DNase I treatment.

Tiling-path DNA microarray

Extracted DNA from each cell line was analyzed by whole genome tiling-path microarrays (SMRT v.2) developed at the British Columbia Cancer Research Centre Array Laboratory.(16) For this array, the whole genome is represented as more than 26,000 overlapping bacterial artificial chromosome (BAC) clones spotted in duplicate with tiling coverage of the human genome. Pooled normal male DNA was used as reference for all microarray experiments. Labelling, hybridization, scanning, and washing of the slides were performed as previously described.(8)

Imaging and analysis of genomic data

Array images were analyzed with GenePix Pro 6.1. A three-step normalization procedure, including locally weighted linear regression (LOWESS) fitting, spatial, and median normalization was used to remove systematic biases.(17) SeeGH software was used to combine duplicate spot data and display log2 signal intensity ratios in relation to genomic locations in the hg18 assembly (NCBI Build 36.1).(18) Data points with standard deviation >0.075 and signal to noise ratio <3 in either channel were removed from each sample.

Two separate algorithms, aCGH-Smooth and DNACopy, were used to detect copy number gains and losses.(19, 20) An alteration was only called when detected concurrently by both algorithms. aCGH-Smooth employs a local search algorithm that uses a maximum likelihood estimation to smooth the observed array CGH values between consecutive breakpoints to a common value.(19) The Lambda and the maximum number of breakpoints in initial pool were set to 6.75 and 100 respectively.(8) DNAcopy employs a circular binary segmentation algorithm that uses a random permutation test to determine the statistical significance of change points.(20) Default settings in DNAcopy were used for our data.

A third algorithm based on moving-average was used for the identification of high-level DNA amplification and presumptive homozygous deletions.(21) The threshold was set at log2 signal intensity ratio >0.8 for high-level amplification or <−0.8 for homozygous deletions. Only regions containing ≥3 overlapping clones with such calls were identified in order to avoid false-positives due to hybridization artefacts.

Gene expression profiling

Total RNA collected from each cell line was analyzed by Agilent Whole Human Genome Microarray 4×44K. This array represents more than 41,000 unique human transcripts. Labelling and hybridizations were performed according to manufacturer’s instructions (Agilent Technologies). Hybridized arrays were scanned using Axon GenePix 4000B and 4200A scanner.

Data analysis of expression profiles

Array images were analyzed using GenePix Pro 6.1. For normalization processing, each background-subtracted intensity value was divided by the median array intensity of each microarray. The median array intensity was calculated based on the background-subtracted intensity value for all spots excluding control type spots on the array. Genes within regions of high-level copy number change were extracted for each cell line. Oral cell lines with no high-level copy number change serve as the baseline for the genes in each alteration region and the mean of each median-normalized intensity value of each probe was compared with those of the cell lines with high-level change.

RESULTS AND DISCUSSION

Cancer cell lines are important model systems for investigating the biology of head and neck cancers. They allow for characterization of a variety of disease phenotypes and also serve as a tool for functional screens. When using such models, it is imperative to determine how closely the model resembles clinical disease. However, genome-wide characterization of most cell lines has not previously been attempted. Characterizing DNA alterations in these cells and determining the contribution of these changes to mRNA expression would provide a very valuable reference point for interpreting experimental results generated using these cell lines. The set of six genomic and six expression profiles has been deposited to Gene Expression Omnibus (GEO) database at NCBI, series accession number GSE16872.

High-level DNA amplification is a common mechanism for driving overexpression of oncogenes, while homozygous deletion is known to inactivate tumour suppressor genes and also contribute to cancer processes.(2123) High resolution genomic analysis delineates the precise boundaries of amplified or deleted regions, thus defining genes that warrant further investigation. Parallel analysis of gene expression data from the same cells helps to differentiate between "driver" and "passenger" genes in a given segmental DNA change, the former being genes that contribute to the cancer phenotype, the latter being altered simply due to its proximity to a "driver" gene.(21, 24) For example, any candidate gene in an amplicon that is not overexpressed is unlikely to represent a "driver" gene. Figure 1 displays a summary of altered regions detected in each oral cell line, while the specific base pair positions are listed in Supplemental tables S1 to S6. Furthermore, as this paper serves as a reference, the copy number status of well known cancer genes obtained from the Cancer Gene Census is catalogued in Table 1.(25) Whole genome copy number karyogram of each cell line and description of each cell line is represented in Supplemental figure S1.

Figure 1.

Figure 1

Summary of copy number alteration in each cell line. Copy number gain is presented as vertical lines on the right side of the chromosome, and vertical lines on the left side indicate copy number loss. High-level copy number change is represented by a star on the right (DNA amplification) or left (homozygous deletion) of the chromosome. Genetic alteration of each cell line is labelled with different colours, SCC-15 (blue), SCC-4 (red), SCC-25 (green), SCC-9 (purple), A-253 (orange), and Cal27 (pink).

Table 1.

Copy number status of cancer genes in the six head and neck cancer cell lines.

Gene Gene Name Gene
ID
Locus SCC-
15
SCC-
4
SCC-
25
SCC-
9
A-
253
Cal27
ABI1 abl-interactor 1 10006 10p12.1
ABL1 v-abl Abelson murine
leukemia viral
oncogene homolog 1
25 9q34.12 + + + +
ABL2 v-abl Abelson murine
leukemia viral
oncogene homolog 2
27 1q24-q25 +
ACSL6 acyl-CoA synthetase
long-chain family
member 6
23305 5q23.3
AF15Q14 AF15q14 protein 57082 15q15.1
AF1Q ALL1-fused gene from
chromosome 1q
10962 1q21 .2 +
AF5q31 ALL1 fused gene from
5q31
27125 5q31.1
AKAP9 A kinase (PRKA)
anchor protein (yotiao)
9
10142 7q21.2 + +
AKT1 v-akt murine thymoma
viral oncogene
homolog 1
207 14q32.33 + +
AKT2 v-akt murine thymoma
viral oncogene
homolog 2
208 19q13.2 +
ALK anaplastic lymphoma
kinase (Ki-1)
238 2p23.2
ALO17 KIAA1618 protein 57714 17q25.3 + +
APC adenomatous
polyposis of the colon
gene
324 5q22.2
ARHGAP26 GTPase regulator
associated with focal
adhesion kinase
pp125(FAK)
23092 5q31.3 +
ARHGEF12 RHO guanine
nucleotide exchange
factor (GEF) 12
(LARG)
23365 11q23.3 + +
ARNT aryl hydrocarbon
receptor nuclear
translocator
405 1q21
ASPSCR1 alveolar soft part
sarcoma chromosome
region, candidate 1
79058 17q25.3 + +
ATF1 activating transcription
factor 1
466 12q13.12 +
ATIC 5-aminoimidazole-4-
carboxamide
ribonucleotide
formyltransferase/IMP
cyclohydrolase
471 2q35
ATM ataxia telangiectasia
mutated
472 11q22.3 +
BCL10 B-cell CLL/lymphoma
10
8915 1p22.3
BCL11A B-cell CLL/lymphoma
11A
53335 2p16.1
BCL11B B-cell CLL/lymphoma
11B (CTIP2)
64919 14q32.2 ++ +
BCL2 B-cell CLL/lymphoma
2
596 18q21.33
BCL3 B-cell CLL/lymphoma
3
602 19q13.31 +
BCL5 B-cell CLL/lymphoma
5
603 17q23.2
BCL6 B-cell CLL/lymphoma
6
604 3q27.3 + + + + + +
BCL7A B-cell CLL/lymphoma
7A
605 12q24.31 + +
BCL9 B-cell CLL/lymphoma
9
607 1q21
BCR breakpoint cluster
region
613 22q11.23 + +
BIRC3 baculoviral IAP repeat-
containing 3
330 11q22.2 + + +
BLM Bloom Syndrome 641 15q26.1
BMPR1A bone morphogenetic
protein receptor, type
IA
657 10q23.2
BRAF v-raf murine sarcoma
viral oncogene
homolog B1
673 7q34
BRCA1 familial breast/ovarian
cancer gene 1
672 17q21.31 +
BRCA2 familial breast/ovarian
cancer gene 2
675 13q13.1 +
BRD4 bromodomain
containing 4
23476 19p13.12
BRIP1 BRCA1 interacting
protein C-terminal
helicase 1
83990 17q23.2
BTG1 B-cell translocation
gene 1, anti-
proliferative
694 12q22
BUB1B BUB1 budding
uninhibited by
benzimidazoles 1
homolog beta (yeast)
701 15q15.1
C12orf9 chromosome 12 open
reading frame 9
93669 12q14.3
C15orf21 chromosome 15 open
reading frame 21
283651 15q21.1
CARD11 caspase recruitment
domain family,
member 11
84433 7p22.2 + + + + + +
CARS cysteinyl-tRNA
synthetase
833 11p15.4
CBFA2T1 core-binding factor,
runt domain, alpha
subunit 2;translocated
to, 1 (ETO)
862 8q21.3 + + +
CBFA2T3 core-binding factor,
runt domain, alpha
subunit 2; translocated
to, 3 (MTG-16)
863 16q24.3
CBFB core-binding factor,
beta subunit
865 16q22.1 +
CBL Cas-Br-M (murine)
ecotropic retroviral
transforming
867 11q23.3 + +
CCDC6 coiled-coil domain
containing 6
8030 10q21.2
CCNB1IP1 enhancer of invasion
10 - fused to HMGA2
57820 14q11.2 + + ++
CCND1 cyclin D1 595 11q13.3 ++ ++ ++ + ++ +
CCND2 cyclin D2 894 12p13.32 +
CCND3 cyclin D3 896 6p21.1 + + +
CDH1 cadherin 1, type 1, E-
cadherin (epithelial)
(ECAD)
999 16q22.1 +
CDH11 cadherin 11, type 2,
OB-cadherin
(osteoblast)
1009 16q21
CDK4 cyclin-dependent
kinase 4
1019 12q14.1 +
CDK6 cyclin-dependent
kinase 6
1021 7q12 ++ +
CDKN2A-
p14ARF
cyclin-dependent
kinase inhibitor 2A--
p14ARF protein
1029 9p21.3
CDKN2A -
p16(INK4a)
cyclin-dependent
kinase inhibitor 2A
(p16(INK4a)) gene
1029 9p21.3
CDX2 caudal type homeo
box transcription factor
2
1045 13q12.2 +
CEBPA CCAAT/enhancer
binding protein
(C/EBP), alpha
1050 19q13.11 +
CEP1 centrosomal protein 1 11064 9q33.2 + + + +
CHEK2 CHK2 checkpoint
homolog (S. pombe)
11200 22q12.1 +
CHIC2 cysteine-rich
hydrophobic domain 2
26511 4q12
CHN1 chimerin (chimaerin) 1 1123 2q31.1
CIC capicua homolog
(Drosophila)
23152 19q13.2 +
CLTC clathrin, heavy
polypeptide (Hc)
1213 17q23.2
CLTCL1 clathrin, heavy
polypeptide-like 1
8218 22q11.21 + +
CMKOR1 chemokine orphan
receptor 1
57007 2q37.3
COL1A1 collagen, type I, alpha
1
1277 17q21.33
COPEB core promoter element
binding protein (KLF6)
1316 10p15.1
COX6C cytochrome c oxidase
subunit VIc
1345 8q22.2 + + + +
CREB1 cAMP responsive
element binding
protein 1
1385 2q33.3
CREB3L2 cAMP responsive
element binding
protein 3-like 2
64764 7q32-q34
CREBBP CREB binding protein
(CBP)
1387 16p13.3 + + +
CTNNB1 catenin (cadherin-
associated protein),
beta 1
1499 3p22.1
CXXC6 Leukemia-associated
protein with a CXXC
domain
80312 10q21.3 +
CYLD familial cylindromatosis
gene
1540 16q12.1 +
DDB2 damage-specific DNA
binding protein 2
1643 11p11.2 + +
DDIT3 DNA-damage-
inducible transcript 3
1649 12q13.3 + +
DDX10 DEAD (Asp-Glu-Ala-
Asp) box polypeptide
10
1662 11q22.3 +
DDX6 DEAD (Asp-Glu-Ala-
Asp) box polypeptide 6
1656 11q23.3 + +
DEK DEK oncogene (DNA
binding)
7913 6p22.3 + +
DUX4 double homeobox, 4 22947 4q35.2
EGFR epidermal growth
factor receptor
(erythroblastic
leukemia viral (v-erb-b)
oncogene homolog,
avian)
1956 7p12.3-
p12.1
++ + + + ++
EIF4A2 eukaryotic translation
initiation factor 4A,
isoform 2
1974 3q27.3 + + + + + +
ELKS ELKS protein 23085 12p13.33 +
ELL ELL gene (11–19
lysine-rich leukemia
gene)
8178 19p13.11
ELN elastin 2006 7q11.23 +
EML4 echinoderm
microtubule associated
protein like 4
27436 2p21
EP300 300 kd E1A-Binding
protein gene
2033 22q13.2
EPS15 epidermal growth
factor receptor
pathway substrate 15
(AF1p)
2060 1p32
ERBB2 v-erb-b2 erythroblastic
leukemia viral
oncogene homolog 2,
neuro/glioblastoma
derived oncogene
homolog (avian)
2064 17q12
ERCC2 excision repair cross-
complementing rodent
repair deficiency,
complementation
group 2 (xeroderma
pigmentosum D)
2068 19q13.32 +
ERCC3 excision repair cross-
complementing rodent
repair deficiency,
complementation
group 3 (xeroderma
pigmentosum group B
complementing)
2071 2q14.3
ERCC4 excision repair cross-
complementing rodent
repair deficiency,
complementation
group 4
2072 16p13.12 + +
ERCC5 excision repair cross-
complementing rodent
repair deficiency,
complementation
group 5 (xeroderma
pigmentosum,
complementation
group G (Cockayne
syndrome))
2073 13q33.1 + +
ERG v-ets erythroblastosis
virus E26 oncogene
like (avian)
2078 21q22.2
ETV1 ets variant gene 1 2115 7p21.2 + + + +
ETV4 ets variant gene 4
(E1A enhancer binding
protein, E1AF)
2118 17q21.31
ETV5 ets variant gene 5 2119 3q37.2 + + + + + +
ETV6 ets variant gene 6
(TEL oncogene)
2120 12p13.2 + +
EVI1 ecotropic viral
integration site 1
2122 3q26.2 + + + +
EWSR1 Ewing sarcoma
breakpoint region 1
(EWS)
2130 22q12.2 +
EXT1 multiple exostoses
type 1 gene
2131 8q24.11 + + + + +
EXT2 multiple exostoses
type 2 gene
2132 11p11.2 + +
FANCA Fanconi anemia,
complementation
group A
2175 16q24.3
FANCC Fanconi anemia,
complementation
group C
2176 9q22.32 + + + +
FANCD2 Fanconi anemia,
complementation
group D2
2177 3p25.3
FANCE Fanconi anemia,
complementation
group E
2178 6p21.31 + + +
FANCF Fanconi anemia,
complementation
group F
2188 11p14.3
FANCG Fanconi anemia,
complementation
group G
2189 9p13.3 ++ +
FBXW7 F-box and WD-40
domain protein 7
(archipelago homolog,
Drosophila)
55294 4q31.3
FCGR2B Fc fragment of IgG,
low affinity IIb, receptor
for (CD32)
2213 1q23 +
FEV FEV protein -
(HSRNAFEV)
54738 2q35
FGFR1 fibroblast growth factor
receptor 1
2260 8p12
FGFR1OP FGFR1 oncogene
partner (FOP)
11116 6q27 + + +
FGFR2 fibroblast growth factor
receptor 2
2263 10q26.12-
q26.13
FGFR3 fibroblast growth factor
receptor 3
2261 4p16.3
FH fumarate hydratase 2271 1q42.1 +
FIP1L1 FIP1 like 1 (S.
cerevisiae)
81608 4q12
FLCN folliculin, Birt-Hogg-
Dube syndrome
201163 17p11.2 + + + + +
FLI1 Friend leukemia virus
integration 1
2313 11q24.3 + +
FLT3 fms-related tyrosine
kinase 3
2322 13q12.2 +
FNBP1 formin binding protein
1 (FBP17)
23048 9q34.11 + + + +
FOXO1A forkhead box O1A
(FKHR)
2308 13q14.11
FOXO3A forkhead box O3A 2309 6q21
FOXP1 forkhead box P1 27086 3p13
FSTL3 follistatin-like 3
(secreted glycoprotein)
10272 19p13.3
FUS fusion, derived from
t(12;16) malignant
liposarcoma
2521 16p11.2 +
FVT1 follicular lymphoma
variant translocation 1
2531 18q21.33
GAS7 growth arrest-specific
7
8522 17p13.1 + + +
GATA2 GATA binding protein
2
2624 3q21.3
GMPS guanine
monphosphate
synthetase
8833 3q25.31 + + + + ++
GNAQ guanine nucleotide
binding protein (G
protein), q
polypeptide
2776 9q21.2 + + +
GNAS guanine nucleotide
binding protein (G
protein), alpha
stimulating activity
polypeptide 1
2778 20q13.32 + + + + + +
GOLGA5 golgi autoantigen,
golgin subfamily a, 5
(PTC5)
9950 14q32.12 ++ +
GOPC golgi associated PDZ
and coiled-coil motif
containing
57120 6q22.1 +
GPHN gephyrin (GPH) 10243 14q23.3 + + + +
HCMOGT-1 sperm antigen
HCMOGT-1
92521 17p11.2 + + + +
HEAB ATP_GTP binding
protein
10978 11q12.1 + + +
HIP1 huntingtin interacting
protein 1
3092 7q11.23 +
HIST1H4I histone 1, H4i (H4FM) 8294 6p22.1 + +
HLF hepatic leukemia factor 3131 17q22
HLXB9 homeo box HB9 3110 7q36
HMGA1 high mobility group AT-
hook 1
3159 6p21.31 + + +
HMGA2 high mobility group AT-
hook 2 (HMGIC)
8091 12q14.3
HNRNPA2B
1
heterogeneous nuclear
ribonucleoprotein
A2/B1
3181 7p15.2 + + + + +
HOXA11 homeo box A11 3207 7p15.2 + + + + +
HOXA13 homeo box A13 3209 7p15.2 + + + + +
HOXA9 homeo box A9 3205 7p15.2 + + + + +
HOXC11 homeo box C11 3227 12q13.13 + +
HOXC13 homeo box C13 3229 12q13.13 + +
HOXD11 homeo box D11 3237 2q31.1
HOXD13 homeo box D13 3239 2q31.1
HRAS v-Ha-ras Harvey rat
sarcoma viral
oncogene homolog
3265 11p15.5
HRPT2 hyperparathyroidism 2 3279 1q21-q31
HSPCA heat shock 90kDa
protein 1, alpha
3320 14q32.31
HSPCB heat shock 90kDa
protein 1, beta
3326 6p21.1 + + +
IDH1 isocitrate
dehydrogenase 1
(NADP+), soluble
3417 2q33.3
IGH@ immunoglobulin heavy
locus
3492 14q32.33 + +
IGKC immunoglobulin kappa
locus
50802 2p11.2
IGL@ immunoglobulin
lambda locus
3535 22q11.1-
q11.2
+ +
IKZF1 IKAROS family zinc
finger 1
10320 7p12.2 + + + + +
IL2 interleukin 2 3558 4q27
IL21R interleukin 21 receptor 50615 16p12.1 + +
IL6ST interleukin 6 signal
transducer (gp130,
oncostatin M
receptor)
3572 5q11.2 +
IRF4 interferon regulatory
factor 4
3662 6p25.3 + +
IRTA1 immunoglobulin
superfamily receptor
translocation
associated 1
83417 1q23.1 +
ITK IL2-inducible T-cell
kinase
3702 5q33.3
JAK2 Janus kinase 2 3717 9p24.1 +
JAK3 Janus kinase 3 3718 19p13.11
JAZF1 juxtaposed with
another zinc finger
gene 1
221895 7p15.2-
p15.1
+ + + + +
KIAA1549 KIAA1549 57670 7q34
KIT v-kit Hardy-Zuckerman
4 feline sarcoma viral
oncogene homolog
3815 4q12
KRAS2 v-Ki-ras2 Kirsten rat
sarcoma 2 viral
oncogene homolog
3845 12p12.1 +
KTN1 kinectin 1 (kinesin
receptor)
3895 14q22.3 + + ++ +
LAF4 lymphoid nuclear
protein related to AF4
3899 2q11.2 +
LASP1 LIM and SH3 protein 1 3927 17q12 +
LCK lymphocyte-specific
protein tyrosine kinase
3932 1p35-
p34.3
+
LCP1 lymphocyte cytosolic
protein 1 (L-plastin)
3936 13q14.13
LHFP lipoma HMGIC fusion
partner
10186 13q13.3
LIFR leukemia inhibitory
factor receptor
3977 5p13.1 + + + + +
LMO1 LIM domain only 1
(rhombotin 1) (RBTN1)
4004 11p15.4
LMO2 LIM domain only 2
(rhombotin-like 1)
(RBTN2)
4005 11p13 + ++ ++
LPP LIM domain containing
preferred translocation
partner in lipoma
4026 3q27.3-
q28
+ + + + + +
LYL1 lymphoblastic
leukemia derived
sequence 1
4066 19p13.13 +
MAF v-maf
musculoaponeurotic
fibrosarcoma
oncogene homolog
4094 16q23.1
MAFB v-maf
musculoaponeurotic
fibrosarcoma
oncogene homolog B
(avian)
9935 20q12 + + + + + +
MALT1 mucosa associated
lymphoid tissue
lymphoma
translocation gene 1
10892 18q21.31
MAML2 mastermind-like 2
(Drosophila)
84441 11q21 + +
MAP2K4 mitogen-activated
protein kinase kinase 4
6416 17p11.2 + + +
MDM2 Mdm2 p53 binding
protein homolog
4193 12q15
MDS1 myelodysplasia
syndrome 1
4197 3q26.2 + + +
MDS2 myelodysplastic
syndrome 2
259283 1p36.12-
p36.11
MECT1 mucoepidermoid
translocated 1
94159 19p13.11 +
MEN1 multiple endocrine
neoplasia type 1 gene
4221 11q13.1 + + + + + +
MET met proto-oncogene
(hepatocyte growth
factor receptor)
4233 7q31.2
MHC2TA MHC class II
transactivator
4261 16p13.13 + + +
MITF microphthalmia-
associated
transcription factor
4286 3p13
MKL1 megakaryoblastic
leukemia
(translocation) 1
57591 22q13.1-
q13.2
MLF1 myeloid leukemia
factor 1
4291 3q25.32 + + + + +
MLH1 E.coli MutL homolog
gene
4292 3p22.3
MLL myeloid/lymphoid or
mixed-lineage
leukemia (trithorax
homolog, Drosophila)
4297 11q23.3 + +
MLLT1 myeloid/lymphoid or
mixed-lineage
leukemia (trithorax
homolog, Drosophila);
translocated to, 1
(ENL)
4298 19p13.3 +
MLLT10 myeloid/lymphoid or
mixed-lineage
leukemia (trithorax
homolog, Drosophila);
translocated to, 10
(AF10)
8028 10p12.31
MLLT2 myeloid/lymphoid or
mixed-lineage
leukemia (trithorax
homolog, Drosophila);
translocated to, 2
(AF4)
4299 4q21.3
MLLT3 myeloid/lymphoid or
mixed-lineage
leukemia (trithorax
homolog, Drosophila);
translocated to, 3
(AF9)
4300 9p21.3
MLLT4 myeloid/lymphoid or
mixed-lineage
leukemia (trithorax
homolog, Drosophila);
translocated to,4
(AF6)
4301 6q27 + + +
MLLT6 myeloid/lymphoid or
mixed-lineage
leukemia (trithorax
homolog, Drosophila);
translocated to, 6
(AF17)
4302 17q21 +
MN1 meningioma (disrupted
in balanced
translocation) 1
4330 22q12.1 +
MPL myeloproliferative
leukemia virus
oncogene,
thrombopoietin
receptor
4352 1p35.1 +
MSF MLL septin-like fusion 10801 17q25 + +
MSH2 mutS homolog 2 (E.
coli)
4436 2p21
MSH6 mutS homolog 6 (E.
coli)
2956 2p16.3
MSI2 musashi homolog 2
(Drosophila)
124540 17q23.2
MUC1 mucin 1,
transmembrane
4582 1q22 +
MUTYH mutY homolog (E. coli) 4595 1p34.1 +
MYC v-myc
myelocytomatosis viral
oncogene homolog
(avian)
4609 8q24.21 + + + + +
MYCL1 v-myc
myelocytomatosis viral
oncogene homolog 1,
lung carcinoma
derived (avian)
4610 1p34.2 +
MYCN v-myc
myelocytomatosis viral
related oncogene,
neuroblastoma derived
(avian)
4613 2p24.3
MYH11 myosin, heavy
polypeptide 11,
smooth muscle
4629 16p13.11 + +
MYH9 myosin, heavy
polypeptide 9, non-
muscle
4627 22q12.3
MYST4 MYST histone
acetyltransferase
(monocytic leukemia) 4
(MORF)
23522 10q22.2 +
NACA nascent-polypeptide-
associated complex
alpha polypeptide
4666 12q13.3 +
NBS1 Nijmegen breakage
syndrome 1 (nibrin)
4683 8q21.3 + + +
NCKIPSD SH3 protein interacting
with Nck, 90 kDa
(ALL1 fused gene from
3p21)
51517 3p21.31
NCOA1 nuclear receptor
coactivator 1
8648 2p23.3
NCOA2 nuclear receptor
coactivator 2 (TIF2)
10499 8q13.3 + +
NCOA4 nuclear receptor
coactivator 4 - PTC3
(ELE1)
8031 10q11.23
NF1 neurofibromatosis type
1 gene
4763 17q11.2
NF2 neurofibromatosis type
2 gene
4771 22q12.2 +
NFKB2 nuclear factor of kappa
light polypeptide gene
enhancer in B-cells 2
(p49/p100)
4791 10q24.32 +
NIN ninein (GSK3B
interacting protein)
51199 14q22.1 + + +
NOTCH1 Notch homolog 1,
translocation-
associated
(Drosophila) (TAN1)
4851 9q34.3 + + + +
NPM1 nucleophosmin
(nucleolar
phosphoprotein B23,
numatrin)
4869 5q35.1 + +
NR4A3 nuclear receptor
subfamily 4, group A,
member 3 (NOR1)
8013 9q31.1 + + +
NRAS neuroblastoma RAS
viral (v-ras) oncogene
homolog
4893 1p13.2 +
NSD1 nuclear receptor
binding SET domain
protein 1
64324 5q35.2-
q35.3
+ +
NTRK1 neurotrophic tyrosine
kinase, receptor, type
1
4914 1q23.1 +
NTRK3 neurotrophic tyrosine
kinase, receptor, type
3
4916 15q25.3
NUMA1 nuclear mitotic
apparatus protein 1
4926 11q13.4 + ++ ++ + +
NUP214 nucleoporin 214kDa
(CAN)
8021 9q34.13 + + + +
NUP98 nucleoporin 98kDa 4928 11p15.4
NUT nuclear protien in testis 256646 15q14
OLIG2 oligodendrocyte
lineage transcription
factor 2 (BHLHB1)
10215 21q22.11 +
OMD osteomodulin 4958 9q22.31 + + + +
PAFAH1B2 platelet-activating
factor acetylhydrolase,
isoform Ib, beta
subunit 30kDa
5049 11q23.3 + +
PALB2 partner and localizer of
BRCA2
79728 16p12.1 + +
PAX3 paired box gene 3 5077 2q36.1
PAX5 paired box gene 5 (B-
cell lineage specific
activator protein)
5079 9p13.2 +
PAX7 paired box gene 7 5081 1p36.13 +
PAX8 paired box gene 8 7849 2q13
PBX1 pre-B-cell leukemia
transcription factor 1
5087 1q23.3 +
PCM1 pericentriolar material
1 (PTC4)
5108 8p22-
p21.3
++
PCSK7 proprotein convertase
subtilisin/kexin type 7
9159 11q23.3 + +
PDE4DIP phosphodiesterase 4D
interacting protein
(myomegalin)
9659 1q21.1 +
PDGFB platelet-derived growth
factor beta
polypeptide
(simian sarcoma viral
(v-sis) oncogene
homolog)
5155 22q13.1
PDGFRA platelet-derived growth
factor, alpha-receptor
5156 4q12
PDGFRB platelet-derived growth
factor receptor, beta
polypeptide
5159 5q32 +
PER1 period homolog 1
(Drosophila)
5187 17p13.1 + + +
PHOX2B paired-like homeobox
2b
8929 4p13 +
PICALM phosphatidylinositol
binding clathrin
assembly protein
(CALM)
8301 11q14.2 + +
PIK3CA phosphoinositide-3-
kinase, catalytic, alpha
polypeptide
5290 3q26.32 + + + +
PIK3R1 phosphoinositide-3-
kinase, regulatory
subunit 1 (alpha)
5295 5q13.1
PIM1 pim-1 oncogene 5292 6p21.2 + + +
PLAG1 pleiomorphic adenoma
gene 1
5324 8q12.1 + + +
PML promyelocytic
leukemia
5371 15q24.1 +
PMS1 PMS1 postmeiotic
segregation increased
1 (S. cerevisiae)
5378 2q32.2
PMS2 PMS2 postmeiotic
segregation increased 2
(S. cerevisiae)
5395 7p22.1 + + + + + +
PNUTL1 peanut-like 1
(Drosophila)
5413 22q11.21 + +
POU2AF1 POU domain, class 2,
associating factor 1
(OBF1)
5450 11q23.1 +
POU5F1 POU domain, class 5,
transcription factor 1
5460 6p21.33 + +
PPARG peroxisome
proliferative activated
receptor, gamma
5468 3p25.2
PRCC papillary renal cell
carcinoma (translocation-
associated)
5546 1q23.1 +
PRDM16 PR domain containing
16
63976 1p36.32 +
PRKAR1A protein kinase, cAMP-
dependent, regulatory,
type I, alpha (tissue
specific extinguisher 1)
5573 17q24.2 +
PRO1073 PRO1073 protein
(ALPHA)
29005 11q13.1 + + + + + +
PRRX1 paired mesoderm
homeo box 1
5396 1q24 .2 + +
PSIP1 PC4 and SFRS1
interacting protein 1
(LEDGF)
11168 9p22.3
PTCH Homolog of Drosophila
Patched gene
5727 9q22.32 + + + +
PTEN phosphatase and
tensin homolog gene
5728 10q23.31
PTPN11 protein tyrosine
phosphatase, non-
receptor type 11
5781 12q24.13 + +
RABEP1 rabaptin, RAB GTPase
binding effector protein
1 (RABPT5)
9135 17p13.2 + + +
RAD51L1 RAD51-like 1 (S.
cerevisiae) (RAD51B)
5890 14q24.1 + + + + +
RANBP17 RAN binding protein
17
64901 5q35.1 + +
RAP1GDS1 RAP1, GTP-GDP
dissociation stimulator
1
5910 4q23
RARA retinoic acid receptor,
alpha
5914 17q21.2 +
RB1 retinoblastoma gene 5925 13q14.2
RBM15 RNA binding motif
protein 15
64783 1p13.3 ++
RECQL4 RecQ protein-like 4 9401 8q24.3 + + + + +
REL v-rel
reticuloendotheliosis
viral oncogene
homolog (avian)
5966 2p16.1
RET ret proto-oncogene 5979 10q11.21
RHOH RAS homolog gene
family, member H
(TTF)
399 4p14 +
ROS1 v-ros UR2 sarcoma
virus oncogene
homolog 1 (avian)
6098 6q22.1
RPL22 ribosomal protein L22
(EAP)
6146 1p36.31 +
RPN1 ribophorin I 6184 3q21.3
RUNX1 runt-related
transcription factor 1
(AML1)
861 21q22.12 +
RUNXBP2 runt-related
transcription factor
binding protein 2
(MOZ/ZNF220)
7994 8p11.21 +
SBDS Shwachman-Bodian-
Diamond syndrome
protein
51119 7q11.21 + +
SDHB succinate
dehydrogenase
complex, subunit B,
iron sulfur (Ip)
6390 1p36.13 +
SDHC succinate
dehydrogenase
complex, subunit C,
integral membrane
protein, 15kDa
6391 1q23.3 +
SDHD succinate
dehydrogenase
complex, subunit D,
integral membrane
protein
6392 11q23.1 +
SET SET translocation 6418 9q33.2 + + + +
SFPQ splicing factor
proline/glutamine
rich(polypyrimidine
tract binding protein
associated)
6421 1p34.3 +
SFRS3 splicing factor,
arginine/serine-rich 3
6428 6p21.31 + + +
SH3GL1 SH3-domain GRB2-
like 1 (EEN)
6455 19p13.3
SIL TAL1 (SCL)
interrupting locus
6491 1p33 +
SLC45A3 solute carrier family
45, member 3
85414 1q32.1 + + +
SMAD4 Homolog of Drosophila
Mothers Against
Decapentaplegic 4
gene
4089 18q21.1
SMARCB1 SWI/SNF related,
matrix associated,
actin dependent
regulator of chromatin,
subfamily b, member 1
6598 22q11.23 + +
SMO smoothened homolog
(Drosophila)
6608 7q32.1
SOCS1 suppressor of cytokine
signaling 1
8651 16p13.13 + + +
SS18 synovial sarcoma
translocation,
chromosome 18
6760 18q11.2 +
SS18L1 synovial sarcoma
translocation gene on
chromosome 18-like 1
26039 20q13.33 + + + + + +
STK11 serine/threonine
kinase 11 gene (LKB1)
6794 19p13.3
STL Six-twelve leukemia
gene
7955 6q22.31 +
SUFU suppressor of fused
homolog (Drosophila)
51684 10q24.32 +
SUZ12 suppressor of zeste 12
homolog (Drosophila)
23512 17q11.2
SYK spleen tyrosine kinase 6850 9q22.2 + + + +
TAF15 TAF15 RNA
polymerase II, TATA
box binding protein
(TBP)-associated
factor, 68kDa
8148 17q12
TAL1 T-cell acute
lymphocytic leukemia
1 (SCL)
6886 1p33 +
TAL2 T-cell acute
lymphocytic leukemia
2
6887 9q31.2 + + + +
TCEA1 transcription
elongation factor A
(SII), 1
6917 8q11.23 + + +
TCF1 transcription factor 1,
hepatic (HNF1)
6927 12q24.31 + +
TCF12 transcription factor 12
(HTF4, helix-loop-helix
transcription factors 4)
6938 15q21.3
TCF3 transcription factor 3
(E2A immunoglobulin
enhancer binding
factors E12/E47)
6929 19p13.3
TCL1A T-cell
leukemia/lymphoma
1A
8115 14q32.13 ++ +
TCL6 T-cell
leukemia/lymphoma 6
27004 14q32.13 ++ +
TFEB transcription factor EB 7942 6p21.1 + + +
TFG TRK-fused gene 10342 3q12.2 +
TFPT TCF3 (E2A) fusion
partner (in childhood
Leukemia)
29844 19q13.42 + + +
TFRC transferrin receptor
(p90, CD71)
7037 3q29 + + + + + +
THRAP3 thyroid hormone
receptor associated
protein 3 (TRAP150)
9967 1p34.3 +
TIF1 transcriptional
intermediary factor 1
(PTC6,TIF1A)
8805 7q34
TLX1 T-cell leukemia,
homeobox 1 (HOX11)
3195 10q24.31 +
TLX3 T-cell leukemia,
homeobox 3
(HOX11L2)
30012 5q35.1 + +
TMPRSS2 transmembrane
protease, serine 2
7113 21q22.3 −−
TNFRSF17 tumor necrosis factor
receptor superfamily,
member 17
608 16p13.13 + + +
TNFRSF6 tumor necrosis factor
receptor superfamily,
member 6 (FAS)
355 10q23.31
TOP1 topoisomerase (DNA) I 7150 20q12 + + + + + +
TP53 tumor protein p53 7157 17p13.1 + + +
TPM3 tropomyosin 3 7170 1q21.3 +
TPM4 tropomyosin 4 7171 19p13.12
TPR translocated promoter
region
7175 1q31.1
TRA@ T cell receptor alpha
locus
6955 14q11.2 + +
TRBC1 T cell receptor beta
locus
6957 7q34
TRD@ T cell receptor delta
locus
6964 14q11.2 + +
TRIM33 tripartite motif-
containing 33
(PTC7,TIF1G)
51592 1p13.2 +
TRIP11 thyroid hormone
receptor interactor 11
9321 14q32.12 + +
TSC1 tuberous sclerosis 1
gene
7248 9q34.13 + + + +
TSC2 tuberous sclerosis 2
gene
7249 16p13.3 + + +
TSHR thyroid stimulating
hormone receptor
7253 14q31.1 + + +
TTL tubulin tyrosine ligase 150465 2q13
USP6 ubiquitin specific
peptidase 6 (Tre-2
oncogene)
9098 17p13.2 + + +
VHL von Hippel-Lindau
syndrome gene
7428 3p25.3
WHSC1 Wolf-Hirschhorn
syndrome candidate
1(MMSET)
7468 4p16.3
WHSC1L1 Wolf-Hirschhorn
syndrome candidate 1-
like 1 (NSD3)
54904 8p12
WRN Werner syndrome
(RECQL2)
7486 8p12
WT1 Wilms tumour 1 gene 7490 11p13 + ++ +
XPA xeroderma
pigmentosum,
complementation
group A
7507 9q22.33 + + + +
XPC xeroderma
pigmentosum,
complementation
group C
7508 3p25.1
ZNF145 zinc finger protein 145
(PLZF)
7704 11q23.2 +
ZNF198 zinc finger protein 198 7750 13q12.11 +
ZNF278 zinc finger protein 278
(ZSG)
23598 22q12.2 +
ZNF331 zinc finger protein 331 55422 19q13.42 + +
ZNF384 zinc finger protein 384
(CIZ/NMP4)
171017 12p13.31 +
ZNF521 zinc finger protein 521 25925 18q11.2 +
ZNF9 zinc finger protein 9 (a
cellular retroviral
nucleic acid binding
protein)
7555 3q21.3
ZNFN1A1 zinc finger protein,
subfamily 1A, 1
(Ikaros)
10320 7p12.2 + + + + +

Symbols: +, low-level copy number gain; ++, high-level copy number gain; −, low-level copy number loss; −−, high-level copy number loss.

It is known that head and neck cancers, like many solid tumours, are a molecularly heterogeneous group where several regions of alteration and genes have been reported at high frequencies but may occur in various combinations in a given tumour. Genetic alterations frequently occurring in tumours indicate an importance for carcinogenesis. Loss of chromosome 3p and 9p are genetic events frequently documented in head and neck tumours, and both have been implicated as one of the earliest changes in oral premalignant lesions and associated with progression risks.(6, 9) All six cell lines revealed segmental losses of chromosome 3p and 9p (detailed regions of genetic alterations are listed in Supplemental Tables S1 to S6). Cal27 revealed only one region of segmental loss of 9p24.1-p23, while the remaining five cell lines exhibited copy number loss at 9p21.3, which contain the tumour suppressor CDKN2A. Genetic loss of chromosome 8p is also an expected change that has been frequently described in clinical specimens.(26, 27) Whole arm 8p loss was found in SCC-15, SCC-9, Cal27, while A253 exhibited a region of homozygous deletion on 8p23.2-p23.2 and a region of high-level DNA amplification on 8p22. Gain of chromosome 8q is also a frequent genetic alteration in head and neck tumours, harbouring the known oncogene MYC.(26) Cell lines A253, SCC-15, SCC-4, SCC-25, and SCC-9 all showed low level gain of 8q24 (MYC). Regions of high-level amplification, including 3q26, 7p11 and 11q13, occur frequently in head and neck tumours.(2630) These regions have been associated with PIK3CA, EGFR, and CCND1 respectively. SCC-4 and SCC-9 did not reveal genetic gain of 3q26.32 whereas the lines A253, SCC-15, and SCC-25 showed low level gains. Cell lines SCC-15 and Cal27 exhibited region of high-level amplification at the EGFR locus, and cell lines SCC-4, SCC-9, and A-253 exhibited low-level copy number gain. Cell lines SCC-9 and Cal27 showed genetic gain of the CCND1 locus, whereas four cell lines A253, SCC-15, SCC-4, and SCC-25 displayed high-level amplification of this locus. Complex alterations on chromosome arm 11q were revealed SCC-15, SCC-4, SCC-25, and A253 (Fig. 2).

Figure 2.

Figure 2

Multiple levels of segmental copy number alterations are detected in six cell lines on chromosome 11q. Each BAC clone is displayed as vertical line representing its genomic coverage. Data points to the left and right of the centre line (purple) represent DNA copy number losses and gains, respectively. Regions of gain are highlighted in red and loss is shaded in green.

One common characteristic of the cell lines is the presence of high-level amplifications, where many of the lines contained multiple regions of high-level DNA amplification. This is similar to tumour genomes where it has been reported that high-level amplification is observed in ~65% of cases, although usually only one or two regions per tumour.(26) High-level amplifications indicating multiple copy numbers and regions of homozygous deletion, where both copies are lost, often result in gene overexpression and underexpression.(21, 22, 31) These types of alteration often arise under selective pressure of genes important for the growth of cancer cells. Therefore we focused our analysis on the expression of genes within these regions. The expression levels of genes within all the regions of high-level DNA amplification and homozygous deletion detected in the six cell lines are presented in Supplemental figure S2. In general regions of homozygous deletion often resulted in lower expression levels for many of the genes within the region, while higher level of expression was often observed in at least one gene within an amplicon compared to cell lines without such amplicon. For example, a common region of high-level amplification of 4.15 Mbp in size on 11p13-p12 was detected in Cal27 and A-253, which contains 20 RefSeq genes (Fig. 3). The expression of 17 of these genes were analyzed, and the gene CD44 has a two-fold increase in expression compared to the other four cell lines without such high-level amplification. Other genes inside this amplicon, PDHX and CAT, also have a 5-fold increase in expression. The expression of CD44 is also among the top five-percentile of all expressed genes in the six cell lines, suggesting that while high-level DNA amplification causes higher expression of the gene, other mechanisms might also cause its high expression in cell lines without such amplicon. However, as there are many ways to regulate gene expression, some of the amplified regions do not show the expected changes in gene expression. Further molecular examinations of epigenetic alterations and sequence analyses would be essential to fully characterize the different molecular aspects of each cell line.

Figure 3.

Figure 3

Integrative analysis of genetic and expression levels of genes within 11p13-p12 amplicon. A, Alignment of chromosome region 11p for six cell lines. Each BAC clone is displayed as vertical line representing its genomic coverage. Minimal region of DNA amplification of A-253 and Cal27 is marked by two black lines and is 4.15 Mbp in size. B, The expression level of 17 genes within this amplicon is analyzed by expression arrays. For each cell line, median-normalized expression values are represented on the y-axis and the corresponding gene is listed on the x-axis.

CONCLUSIONS

Head and neck cancer cell lines are important model systems for investigating head and neck cancer biology. Comprehensive characterization of their genetic alterations is useful for the selection and interpretation of studies using cell lines. Our data presents the first comprehensive catalogue of copy number and expression alterations in six commonly used and easily obtainable oral cancer cell lines, thus providing a good resource for researchers to select and use these cell lines in experiments.

Supplementary Material

Supp Fig s1. Supplemental figure 1.

Whole genome copy number karyogram of each cell line. SeeGH software was used to plot the log2 signal intensity ratios of cell line sample versus normal reference male genomic DNA at each BAC. Discussion of each cell line is described underneath the figure.

Supp Fig s2. Supplemental figure 2.

Scatter plots of median-normalized expression values for genes within regions of high-level copy number change. The median-normalized expression values are plotted on the y-axis, and the corresponding gene for each probe on the Agilent Whole Human Genome Microarray 4×44K microarray are plotted on the x-axis. The regions of high-level copy number alteration are detailed in Supplemental Tables S1 to S6.

Supp Table s1
Supp Table s2
Supp Table s3
Supp Table s4
Supp Table s5
Supp Table s6

Acknowledgements

This work was supported by grants from the National Institute of Dental and Craniofacial Research/National Institutes of Health (R01 DE17013), Canadian Institutes of Health Research, and funds from the Pacific Otolaryngology Foundation. IFLT is supported by scholarships from the Canadian Institutes of Health Research (CIHR) and the Michael Smith Foundation for Health Research (MSFHR). We would like to acknowledge Drs Wan Lam and Timon Buys for helpful discussion.

REFERENCES

  • 1.Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin. 2005;55(2):74–108. doi: 10.3322/canjclin.55.2.74. [DOI] [PubMed] [Google Scholar]
  • 2.Lin CJ, Grandis JR, Carey TE, et al. Head and neck squamous cell carcinoma cell lines: established models and rationale for selection. Head Neck. 2007;29(2):163–188. doi: 10.1002/hed.20478. [DOI] [PubMed] [Google Scholar]
  • 3.Martin CL, Reshmi SC, Ried T, et al. Chromosomal imbalances in oral squamous cell carcinoma: examination of 31 cell lines and review of the literature. Oral Oncol. 2008;44(4):369–382. doi: 10.1016/j.oraloncology.2007.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Singh B. Molecular pathogenesis of head and neck cancers. J Surg Oncol. 2008;97(8):634–639. doi: 10.1002/jso.21024. [DOI] [PubMed] [Google Scholar]
  • 5.Califano J, van der Riet P, Westra W, et al. Genetic progression model for head and neck cancer: implications for field cancerization. Cancer Res. 1996;56(11):2488–2492. [PubMed] [Google Scholar]
  • 6.Mao L, Lee JS, Fan YH, et al. Frequent microsatellite alterations at chromosomes 9p21 and 3p14 in oral premalignant lesions and their value in cancer risk assessment. Nat Med. 1996;2(6):682–685. doi: 10.1038/nm0696-682. [DOI] [PubMed] [Google Scholar]
  • 7.Rosin MP, Cheng X, Poh C, et al. Use of allelic loss to predict malignant risk for low-grade oral epithelial dysplasia. Clin Cancer Res. 2000;6(2):357–362. [PubMed] [Google Scholar]
  • 8.Tsui IF, Rosin MP, Zhang L, Ng RT, Lam WL. Multiple aberrations of chromosome 3p detected in oral premalignant lesions. Cancer Prev Res (Phila Pa) 2008;1(6):424–429. doi: 10.1158/1940-6207.CAPR-08-0123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mao L, Hong WK, Papadimitrakopoulou VA. Focus on head and neck cancer. Cancer Cell. 2004;5(4):311–316. doi: 10.1016/s1535-6108(04)00090-x. [DOI] [PubMed] [Google Scholar]
  • 10.Akervall JA, Michalides RJ, Mineta H, et al. Amplification of cyclin D1 in squamous cell carcinoma of the head and neck and the prognostic value of chromosomal abnormalities and cyclin D1 overexpression. Cancer. 1997;79(2):380–389. [PubMed] [Google Scholar]
  • 11.Nakahara Y, Shintani S, Mihara M, Ueyama Y, Matsumura T. High frequency of homozygous deletion and methylation of p16(INK4A) gene in oral squamous cell carcinomas. Cancer Lett. 2001;163(2):221–228. doi: 10.1016/s0304-3835(00)00699-6. [DOI] [PubMed] [Google Scholar]
  • 12.Jarvinen AK, Autio R, Kilpinen S, et al. High-resolution copy number and gene expression microarray analyses of head and neck squamous cell carcinoma cell lines of tongue and larynx. Genes Chromosomes Cancer. 2008;47(6):500–509. doi: 10.1002/gcc.20551. [DOI] [PubMed] [Google Scholar]
  • 13.Rheinwald JG, Beckett MA. Tumorigenic keratinocyte lines requiring anchorage and fibroblast support cultures from human squamous cell carcinomas. Cancer Res. 1981;41(5):1657–1663. [PubMed] [Google Scholar]
  • 14.Gioanni J, Fischel JL, Lambert JC, et al. Two new human tumor cell lines derived from squamous cell carcinomas of the tongue: establishment, characterization and response to cytotoxic treatment. Eur J Cancer Clin Oncol. 1988;24(9):1445–1455. doi: 10.1016/0277-5379(88)90335-5. [DOI] [PubMed] [Google Scholar]
  • 15.Giard DJ, Aaronson SA, Todaro GJ, et al. In vitro cultivation of human tumors: establishment of cell lines derived from a series of solid tumors. J Natl Cancer Inst. 1973;51(5):1417–1423. doi: 10.1093/jnci/51.5.1417. [DOI] [PubMed] [Google Scholar]
  • 16.Ishkanian AS, Malloff CA, Watson SK, et al. A tiling resolution DNA microarray with complete coverage of the human genome. Nat Genet. 2004;36(3):299–303. doi: 10.1038/ng1307. [DOI] [PubMed] [Google Scholar]
  • 17.Khojasteh M, Lam WL, Ward RK, MacAulay C. A stepwise framework for the normalization of array CGH data. BMC Bioinformatics. 2005;6:274. doi: 10.1186/1471-2105-6-274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Chi B, deLeeuw RJ, Coe BP, Ng RT, MacAulay C, Lam WL. MD-SeeGH: a platform for integrative analysis of multi-dimensional genomic data. BMC Bioinformatics. 2008;9:243. doi: 10.1186/1471-2105-9-243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Jong K, Marchiori E, Meijer G, Vaart AV, Ylstra B. Breakpoint identification and smoothing of array comparative genomic hybridization data. Bioinformatics. 2004;20(18):3636–3637. doi: 10.1093/bioinformatics/bth355. [DOI] [PubMed] [Google Scholar]
  • 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(4):557–572. doi: 10.1093/biostatistics/kxh008. [DOI] [PubMed] [Google Scholar]
  • 21.Lockwood WW, Chari R, Coe BP, et al. DNA amplification is a ubiquitous mechanism of oncogene activation in lung and other cancers. Oncogene. 2008;27(33):4615–4624. doi: 10.1038/onc.2008.98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hahn SA, Schutte M, Hoque AT, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science. 1996;271(5247):350–353. doi: 10.1126/science.271.5247.350. [DOI] [PubMed] [Google Scholar]
  • 23.Snijders AM, Schmidt BL, Fridlyand J, et al. Rare amplicons implicate frequent deregulation of cell fate specification pathways in oral squamous cell carcinoma. Oncogene. 2005;24(26):4232–4242. doi: 10.1038/sj.onc.1208601. [DOI] [PubMed] [Google Scholar]
  • 24.Haber DA, Settleman J. Cancer: drivers and passengers. Nature. 2007;446(7132):145–146. doi: 10.1038/446145a. [DOI] [PubMed] [Google Scholar]
  • 25.Futreal PA, Coin L, M Marshall, et al. A census of human cancer genes. Nat Rev Cancer. 2004;4(3):177–183. doi: 10.1038/nrc1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Baldwin C, Garnis C, Zhang L, Rosin MP, Lam WL. Multiple microalterations detected at high frequency in oral cancer. Cancer Res. 2005;65(17):7561–7567. doi: 10.1158/0008-5472.CAN-05-1513. [DOI] [PubMed] [Google Scholar]
  • 27.Smeets SJ, Braakhuis BJ, Abbas S, et al. Genome-wide DNA copy number alterations in head and neck squamous cell carcinomas with or without oncogene-expressing human papillomavirus. Oncogene. 2006;25(17):2558–2564. doi: 10.1038/sj.onc.1209275. [DOI] [PubMed] [Google Scholar]
  • 28.Redon R, Muller D, Caulee K, Wanherdrick K, Abecassis J, du Manoir S. A simple specific pattern of chromosomal aberrations at early stages of head and neck squamous cell carcinomas: PIK3CA but not p63 gene as a likely target of 3q26-qter gains. Cancer Res. 2001;61(10):4122–4129. [PubMed] [Google Scholar]
  • 29.Huang X, Godfrey TE, Gooding WE, McCarty KS, Jr, Gollin SM. Comprehensive genome and transcriptome analysis of the 11q13 amplicon in human oral cancer and synteny to the 7F5 amplicon in murine oral carcinoma. Genes Chromosomes Cancer. 2006;45(11):1058–1069. doi: 10.1002/gcc.20371. [DOI] [PubMed] [Google Scholar]
  • 30.Huang X, Gollin SM, Raja S, Godfrey TE. High-resolution mapping of the 11q13 amplicon and identification of a gene, TAOS1, that is amplified and overexpressed in oral cancer cells. Proc Natl Acad Sci U S A. 2002;99(17):11369–11374. doi: 10.1073/pnas.172285799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Albertson DG. Gene amplification in cancer. Trends Genet. 2006;22(8):447–455. doi: 10.1016/j.tig.2006.06.007. [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Supp Fig s1. Supplemental figure 1.

Whole genome copy number karyogram of each cell line. SeeGH software was used to plot the log2 signal intensity ratios of cell line sample versus normal reference male genomic DNA at each BAC. Discussion of each cell line is described underneath the figure.

Supp Fig s2. Supplemental figure 2.

Scatter plots of median-normalized expression values for genes within regions of high-level copy number change. The median-normalized expression values are plotted on the y-axis, and the corresponding gene for each probe on the Agilent Whole Human Genome Microarray 4×44K microarray are plotted on the x-axis. The regions of high-level copy number alteration are detailed in Supplemental Tables S1 to S6.

Supp Table s1
Supp Table s2
Supp Table s3
Supp Table s4
Supp Table s5
Supp Table s6

RESOURCES