Skip to main content
Erschienen in: Virchows Archiv 3/2009

Open Access 01.09.2009 | Original Article

High resolution analysis of DNA copy-number aberrations of chromosomes 8, 13, and 20 in gastric cancers

verfasst von: Tineke E. Buffart, Nicole C. T. van Grieken, Marianne Tijssen, Jordy Coffa, Bauke Ylstra, Heike I. Grabsch, Cornelis J. H. van de Velde, Beatriz Carvalho, Gerrit A. Meijer

Erschienen in: Virchows Archiv | Ausgabe 3/2009

Abstract

DNA copy-number gains of chromosomes 8q, 13q, and 20q are frequently observed in gastric cancers. Moreover gain of chromosome 20q has been associated with lymph node metastasis. The aim of this study was to correlate DNA copy-number changes of individual genes on chromosomes 8q, 13q, and 20q in gastric adenocarcinomas to clinicopathological data. DNA isolated from 63 formalin-fixed and paraffin-embedded gastric adenocarcinoma tissue samples was analyzed by whole-genome microarray comparative genomic hybridization and by multiplex ligation-dependent probe amplification (MLPA), targeting 58 individual genes on chromosomes 8, 13, and 20. Using array comparative genomic hybridization, gains on 8q, 13q, and 20q were observed in 49 (77.8%), 25 (39.7%), and 49 (77.8%) gastric adenocarcinomas, respectively. Gain of chromosome 20q was significantly correlated with lymph node metastases (p = 0.05) and histological type (p = 0.02). MLPA revealed several genes to be frequently gained in DNA copy number. The oncogene c-myc on 8q was gained in 73% of the cancers, while FOXO1A and ATP7B on 13q were both gained in 28.6% of the cases. Multiple genes on chromosome 20q showed gains in more than 60% of the cancers. DNA copy-number gains of TNFRSF6B (20q13.3) and ZNF217 (20q13.2) were significantly associated with lymph node metastasis (p = 0.02) and histological type (p = 0.02), respectively. In summary, gains of chromosomes 8q, 13q, and 20q in gastric adenocarcinomas harbor DNA copy-number gains of known and putative oncogenes. ZNF217 and TNFRSF6B are associated with important clinicopathological variables, including lymph node status.

Introduction

Gastric cancer is a major health problem and ranks second as a cause of cancer death worldwide [1]. The only possible curative treatment is complete surgical resection. One of the hallmarks of solid cancers, including gastric cancer, is chromosomal instability leading to gains and losses of parts of, or even whole chromosomes [2]. The mechanisms underlying this instability phenotype in gastric cancers are still poorly understood. Patterns of DNA copy-number aberrations can be analyzed at high resolution by array comparative genomic hybridization (CGH). Gains of chromosomes 3q, 7p, 7q, 8q, 13q, 17q, and 20q and losses of chromosomes 4q, 5q, 6q, 9p, 17p, and 18q have been consistently described in gastric cancer studies using CGH or array CGH analysis [311]. In addition, gains of chromosomes 7q, 8q, 9q, 11q, 13q, and 20q and losses of chromosomes 4p, 5q, 6, 9p, 17p, and 18q can already be detected in gastric cancer precursor lesions with direct malignant potential, indicating these as early events in the pathogenesis of gastric cancer [1216]. Results from array CGH studies have shown that patterns of chromosomal aberrations can be correlated with clinicopathological variables. In previous studies, we have shown different patterns of chromosomal instability to correlate with lymph node status and with age of onset of the disease [17, 18].
DNA copy-number gains of chromosomes 8, 13, and 20 have been described to play an important role in colorectal adenoma to carcinoma progression [19, 20]. Also in gastric adenomas and adenocarcinomas, gains of these chromosomes have frequently been detected, indicating the importance of genetic events on these chromosomes for gastric cancer pathogenesis. Moreover, gain of chromosome 20q has been described to correlate with lymph node metastasis in gastric cancer and poor clinical outcome in colorectal cancer [3, 21, 22]. The most widely used array CGH platforms, so far, use spotted bacterial artificial chromosomes (BACs), which means that the probe sequences on the arrays cover up to 1 kb of DNA. This approach is very suitable for detecting patterns of DNA copy-number changes but has limitations in pinpointing individual genes whose normal function is disrupted due to these chromosomal aberrations. Multiplex ligation-dependent probe amplification (MLPA) [23] allows to determine in a single experiment DNA copy-number ratios of up to 40 individual genes and can be used to zoom in on chromosomal areas that show aberrations with array CGH [24, 25]. The aim of the present study is a detailed analysis of DNA copy-number changes of individual genes within gained areas on chromosomes 8, 13, and 20 in gastric adenocarcinomas and to correlate gene specific alterations to clinicopathological data.

Materials and methods

Materials and DNA isolation

DNA of 63 formalin-fixed and paraffin-embedded (FFPE) gastric adenocarcinomas of which 53 were obtained from Leeds (Leeds University Hospital, UK) and 10 were obtained from the Dutch D1/D2 trial [26], was isolated as previously described [17] using a commercial available DNA isolation kit (QIAmp DNA microkit, Qiagen, Hilden, Germany). Briefly, areas of at least 70% tumor cells were demarcated on a 4-µm haematoxylin- and eosin-stained tissue section. Adjacent serial sections of 10 µm were cut, and after deparaffination, the tumor tissue was macrodissected using a needle. After an overnight incubation with sodium thiocyanate (1 M) at 37°C, followed by proteinase K treatment, DNA was extracted. DNA concentrations were measured on a Nanodrop ND-1000 spectrophotometer (Isogen, IJsselstein, The Netherlands), and DNA quality was assessed by isothermal amplification as previously described [27]. Only DNA of excellent, good, and intermediate quality was used for further analysis.

DNA labeling and array CGH)

DNA labeling and array CGH were essentially performed as previously described [17, 28]. In short, tumor and normal DNA were differentially labeled using random priming (Bioprime DNA Labeling System; Invitrogen, Breda, The Netherlands) and hybridized on a BAC array containing approximately 5,000 clones printed in triplicate, consisting of the Sanger BAC clone set with an average resolution along the whole genome of 1.0 Mb (http://​www.​ensembl.​org/​Homo_​sapiens/​cytoview), the OncoBac set (http://​informa.​bio.​caltech.​edu/​Bac_​onc.​html), containing approximately 600 clones corresponding to 200 cancer-related genes, and selected clones of interest obtained from the Children’s Hospital Oakland Research Institute (CHORI) to fill any gaps larger than 1 Mb on chromosome 6 and to have full-coverage contigs of regions on chromosomes 8, 11, 13, and 20. All clones were printed on Codelink™ slides (Amersham BioSciences, Roosendaal, The Netherlands) at a concentration of 1 μg/μl in 150-mM sodium phosphate, pH 8.5, using an OmniGrid 100 microarrayer (Genomic Solutions, Ann Arbor, MI, USA) equipped with SMP3 pins (TeleChem International, Sunnyvale, CA, USA), and processed according to the manufacturer’s protocol.
After hybridization, images of the arrays were acquired by scanning (Microaray scanner G2505B; Agilent technologies, Palo Alto, USA), and spot analysis and quality control were automatically performed using BlueFuse 3.4 software (BlueGnome, Cambridge, UK). When the BlueFuse quality flag was below one or the confidence value was below 0.1, spots were excluded from further analysis. The log2 tumor to normal fluorescence ratio was calculated for each spot and normalized against the mode of the ratios of all autosomes. The package CGH call was used for data segmentation and defining copy-number gains and losses of each clone in the array CGH profile [29].

Multiplex ligation-dependent probe amplification

MLPA was performed, as previously described, [24] using two different probe mixes. One probemix contained 38 probes representing 31 different genes on chromosome 20 and 10 control probes located on chromosomes 2, 3, 4, 5, 12, and 16. The second probemix contained 11 probes on chromosome 8, 12 probes on chromosome 13, 16 probes on chromosome 20 representing 14 different genes, and 8 control probes located on chromosomes 2, 4, 12, and 16. Some genes on chromosome 20 are present in both probe mixes, leaving 35 different genes on chromosome 20 by combining these two probe mixes. DNA of the cell line HT29, showing a gain on chromosomes 8, 13, and 20, was used as positive control. A human pool of DNA isolated from blood of 36 healthy individuals and a pool of DNA isolated of 30 normal gastric and colon mucosa, spleen, liver, and kidney tissue samples (FFPE), were used as normal controls.
Of each sample, approximately 100 ng of DNA in a volume of 5 µl was denaturated at 98°C for 5 min. A mixture of 1.5-µl salsa probes (1–4 fmol of each short synthetic probe oligonucleotide and each phage M13-derived long probe oligonucleotide in TE (10 mM Tris-HCl, pH 8.2, 1 mM ethylenediamine tetraacetic acid (EDTA))) and 1.5 µl of MLPA buffer (1.5 M KCl, 300 mM Tris-HCl, pH 8.5, 1 mM EDTA) was added. The mixture was heated for 1 min at 95°C followed by 16 h of incubation at 60°C to allow the MLPA hemipobes to hybridize. Next, 32 µl of ligase-65 mixture (dilution buffer containing 2.6 mM MgCl2, 5 mM Tris-HCl, pH 8.5, 0.013% non-ionic detergents, 0.2 mM of nicotinamide adenine dinucleotide (NAD), and 1 U of ligase-65 enzyme) was added to each sample for ligation of hybridized hemiprobes during a 10–15 min of incubation at 54°C, followed by a 5 min of incubation at 98°C to inactivate the ligase.
Polymerase chain reaction (PCR) was performed with 10 µl of polymerase mixture containing the PCR primers (10 pmol), dNTPs (2.5 nmol), and 2.5 U Taq polymerase (promega), 4 µl of PCR buffer (2.6 mM MgCl2, 5 mM Tris-HCl, pH 8.5, 0.013% non-ionic detergents, 0.2 mM NAD), 26 µl of water and 10 µl of MLPA ligation reaction.

Multiplex ligation-dependent probe amplification data analysis

Analysis of the MLPA PCR products for each gene was performed on an ABI 3100 capillary sequencer (Applied Biosystems, Warrington, UK) in a mixture of 8.5 µl of deionized formamide (Applied Biosystems, Warrington, UK), 1 µl of PCR product and 0.5 µl marker including a ROX-labeled internal size standard (ROX-500 Genescan; Applied Biosystems, Warrington, UK). Data analysis was performed using the MLPAnalyzer version 8.0 (http://​www.​mlpa.​com/​coffalyser/​) [30]. For each tumor, peak heights for every probe were derived from the ABI output and median peak heights of at least two different ligation reactions and three different PCR reactions were calculated. For each sample, tumor to normal DNA copy-number ratios was calculated per probe by dividing the median peak heights in the tumor tissue by the median peak heights in the reference DNA. All ratios were normalized by setting the median of the tumor to reference DNA copy-number ratios of the control genes in the probe mixture to 1.0. When multiple probes were present for one gene, the mean value of the probes was calculated and used for further analysis. Tumor to normal ratios below 0.7 and above 1.3 was considered as a loss or gain, respectively. TMEV software 3.1 (http://​www.​tigr.​org/​) was used to present descriptive data.

Statistical analysis

Box and scatter plots were used to present descriptive statistics. Chi-square test was used to evaluate associations of DNA copy-number gain of chromosomes 8q, 13q, and 20q with clinicopathological variables. t Test was used to evaluate differences in DNA copy-number aberrations and age of the patients. Mann-Whitney U test and Kruskal-Wallis H test were used to evaluate differences in DNA copy-number changes of each gene between lymph node status and histological tumor type according to the Laurén classification [31], respectively. Correlation coefficients between the log2 ratios for the array CGH and MLPA analysis were obtained by Spearman correlation (SPSS 12.0.1 for Windows; SPSS Inc. Chicago, IL, USA). A threshold of 0.05 for significance was used.

Results

Array CGH analysis and correlation with lymph node status and histological type

Of the 63 gastric adenocarcinomas analyzed by array CGH, 49 (77.8%) showed gains on chromosome 8, 25 (39.7%) showed gains on chromosome 13, and 49 (77.8%) showed gains on chromosome 20. Concurrent gains of chromosomes 8 and 13, 8 and 20, and 13 and 20 were observed in four (6.3%), 24 (38.1%), and three (4.8%) of the gastric adenocarinomas, respectively. Concurrent gains of chromosomes 8, 13, and 20 were observed in 17 (27%) gastric adenocarcinomas. Gain of chromosome 20q was significantly correlated with lymph node status (p = 0.05) and histological type of the tumor (p = 0.02), being more common in lymph node-positive gastric cancers (85%) than in lymph node-negative gastric cancers (64%) and more common in intestinal-type (87%) and mixed-type (88%) gastric cancers than in diffuse-type (53%) gastric cancers. No significant (NS) correlation was found between 20q gain and age and gender of the patients and tumor size. No correlation of 8q gain or 13q gain with clinicopathological variables was found. An overview of the frequencies of copy-number gains on chromosomes 8q, 13q, and 20q detected by array CGH analysis is given in Table 1. An overview of patient and tumor characteristics is given in Table 2.
Table 1
An overview of DNA copy number gains of chromosomes 8q, 13q and 20q detected by array CGH in 63 gastric adenocarcinomas, and the correlation to lymph node status and histological type of the tumor
Chromosome
Status
All cases
Lymph node status
P value
Histological type
P value
LN0
LN1
Intestinal
Diffuse
Mixed
n = 63
n = 22
n = 41
n = 38
n = 17
n = 8
8q
Gain
49 (77.8%)
18 (81.8%)
31 (75.6%)
0.57
29 (76.3%)
13 (76.5%)
7 (87.5%)
0.78
 
No gain
14 (22.2%)
4 (18.2%)
10 (24.4%)
9 (23.7%)
4 (23.5%)
1 (12.5%)
13q
Gain
25 (39.7%)
10 (45.5%)
15 (36.6%)
0.49
19 (50.0%)
4 (23.5%)
2 (25.0%)
0.12
 
No gain
38 (60.3%)
12 (54.5%)
26 (63.4%)
19 (50.0%)
13 (76.5%)
6 (75.0%)
20q
Gain
49 (77.8%)
14 (63.6%)
35 (85.4%)
0.048
33 (86.8%)
9 (52.9%)
7 (87.5%)
0.016
 
No gain
14 (22.2%)
8 (36.4%)
6 (14.6%)
5 (13.2%)
8 (47.1%)
1 (12.5%)
8q + 13q + 20q
Gain
17 (27.0%)
6 (27.2%)
11 (26.8%)
0.97
13 (34.2%)
2 (11.8%)
2 (25.0%)
0.22
 
No gain
46 (73.0%)
16 (72.7%)
30 (73.2)
25 (65.8%)
15 (88.2%)
6 (75.0%)
8q + 13q
Gain
4 (6.3%)
3 (13.6%)
1 (2.4%)
0.082
2 (5.3%)
2 (11.8%)
0 (0.0%)
0.48
 
No gain
59 (93.7%)
19 (86.4%)
40 (97.6%)
36 (94.7%)
15 (88.2%)
8 (100.0%)
8q + 20q
Gain
24 (38.1%)
8 (36.4%)
16 (39.0%)
0.84
14 (36.8%)
6 (35.3%)
4 (50.0%)
0.76
 
No gain
39 (61.9%)
14 (63.6%)
25 (61.0%)
24 (63.2%)
11 (64.7%)
4 (50.0%)
13q + 20q
Gain
3 (4.8%)
0 (0.0%)
3 (7.3%)
0.19
3 (7.9%)
0 (0.0%)
0 (0.0%)
0.36
 
No gain
60 (95.2%)
22 (100.0%)
38 (92.7%)
35 (92.1%)
17 (100.0%)
8 (100.0%)
Gain of 20q is significantly correlated to lymph node status and histological tumor type (in bold)
LN0 lymph node-negative, LN1 lymph node-positive
Table 2
Overview of patient and tumor characteristics of the 63 gastric adenocarcinomas analyzed in the study
Tumor ID
Gender
Age
Histological type
T status
N status
Tumor ID
Gender
Age
Histological type
T status
N status
1
Female
78
Intestinal
T3
N1
33
Male
75
Intestinal
T2
N3
2
Female
81
Diffuse
T3
N1
34
Male
61
Intestinal
T3
N3
3
Female
54
Intestinal
T2
N0
35
Female
82
Diffuse
T3
N1
4
Male
73
Intestinal
T3
N1
36
Male
74
Mixed
T2
N1
5
Male
81
Mixed
T2
N2
37
Female
87
Diffuse
T3
N2
6
Male
58
Intestinal
T2
N1
38
Female
78
Intestinal
T1
N0
7
Female
72
Intestinal
T2
N0
39
Male
78
Diffuse
T2
N0
8
Male
86
Intestinal
T2
N2
40
Female
84
Intestinal
T2
N0
9
male
71
Intestinal
T3
N0
41
Male
85
Diffuse
T3
N2
10
Female
75
Intestinal
T4
N1
42
Male
62
Diffuse
T3
N3
11
Female
75
Intestinal
T3
N2
43
Male
68
Intestinal
T3
N1
12
Male
64
Mixed
T3
N1
44
Male
58
Intestinal
T4
N1
13
Male
81
Intestinal
T2
N2
45
Male
61
Diffuse
T3
N1
14
Male
63
Intestinal
T2
N0
46
Female
58
Intestinal
T3
N1
15
Male
63
Intestinal
T3
N1
47
Male
81
Intestinal
T3
N1
16
Male
68
Mixed
T2
N0
48
Female
65
Intestinal
T2
N1
17
Female
72
Intestinal
T2
N1
49
Female
65
Intestinal
T2
N1
18
Male
73
Intestinal
T2
N2
50
Male
82
Intestinal
T1
N0
19
Male
71
Intestinal
T3
N2
51
Female
74
Mixed
T1
N0
20
Female
67
Intestinal
T2
N0
52
Male
72
Diffuse
T3
N2
21
Male
67
Diffuse
T3
N1
53
Female
60
Diffuse
T3
N0
22
Female
64
Intestinal
T3
N2
54
Male
47
Diffuse
T2
N0
23
Female
72
Diffuse
T3
N2
55
Female
74
Diffuse
T2
N0
24
Male
74
Intestinal
T2
N1
56
Male
65
Mixed
T2
N2
25
Male
58
Intestinal
T3
N3
57
Female
49
Intestinal
T1
N0
26
Male
71
Intestinal
T1
N0
58
Male
74
Intestinal
T1
N0
27
Female
68
Mixed
T3
N1
59
Male
58
Intestinal
T1
N0
28
Male
74
Intestinal
T2
N0
60
Male
53
Diffuse
T2
N0
29
Male
57
Mixed
T3
N1
61
Male
61
Diffuse
T2
N1
30
Male
69
Diffuse
T3
N1
62
Male
69
Diffuse
T2
N1
31
Male
67
Intestinal
T2
N0
63
Male
56
Intestinal
T2
N0
32
Female
71
Intestinal
T2
N1
      

Multiplex ligation-dependent probe amplification analysis of DNA copy-number aberrations and correlation with lymph node status and histological type

All 63 gastric adenocarcinomas analyzed by array CGH were analyzed by MLPA. An overview of the DNA copy-number ratios of all individual genes on chromosomes 8, 13, and 20 in all tumor samples is presented as a heatmap in Fig. 1.
Gains of genes on chromosome 8q were observed in 9.5%–73.0% of the gastric adenocarcinomas, with the highest frequencies of gains observed in c-myc (73.0%). Gains of genes on chromosome 13q were detected in 11.1%–28.6%, with the highest frequencies of gains observed in FOXO1A and ATP7B (both 28.6%). Frequent DNA copy-number gains of multiple genes on chromosome 20q were observed. Gain of TOP1, PTPN1, CYP24A1, and GATA5 was observed in more than 60% of the gastric adenocarcinomas, and gain of BCL2L1, MYBL2, STX16, and GNAS was observed in more than 70% of the gastric adenocarcinomas. When correlating gene-specific copy-number status to lymph node status, EIF3S6 (p = 0.04), located on chromosome 8q22 and TNFRSF6B (p = 0.02), located on chromosome 20q13.3, were significantly different between lymph node-positive and lymph node-negative gastric adenocarcinomas. Although Mann-Whitney U test yielded a significant difference in copy-number of the gene EIF3S6 between lymph node-positive and lymph node-negative gastric adenocarcinomas, mean copy-number ratio of both carcinoma groups were within the normal copy-number range (DNA copy-number ratio between 0.7 and 1.3). Lymph node-positive gastric adenocarcinomas showed gain (DNA copy-number ratio >1.3) of TNFRSF6B while lymph node negative gastric adenocarcinomas showed normal copy-number ratios of this gene (Fig. 2). When correlating gene-specific copy-number ratio to histological tumor type, Kruskall-Wallis H test yielded five significant genes. Mean copy-number ratios of FBXO25 (p = 0.01), located on 8p23.3, RASL11A (p = 0.03), located on 13q12.2 and CDX2 (p = 0.03), located on 13q12, were within the normal copy-number range for all three tumor types. Mean copy-number ratio of ZNF217 (p = 0.02), located on 20q13.2, showed gain in intestinal and mixed histological gastric cancer types versus normal in the diffuse histological types (Fig. 3). STX16 (p = 0.03), located on 20q13.32, showed mean copy-number gain in all three tumor types, but higher mean copy-number ratios in the intestinal and mixed types of gastric carcinomas compared with diffuse-type gastric carcinomas (1.90, 2.03, and 1.47, respectively). A detailed overview of the frequencies of gains per gene and mean copy-number ratio per tumor group, i.e., lymph node-positive versus lymph node-negative gastric adenocarcinomas and intestinal- versus diffuse- versus mixed-type gastric carcinomas are given in Table 3.
Table 3
A detailed overview of the frequencies of DNA copy number gains per gene, determined by MLPA analysis and mean DNA copy number ratio and range, per tumor group, i.e., lymph node-positive versus lymph node-negative gastric adenocarcinomas and intestinal- versus diffuse- versus mixed-type gastric carcinomas
Gene
Location
Gains
LN-negative
LN-positive
p value
Intestinal
Diffuse
Mixed
p value
FBXO25
8p23.3
1.6%
1.04 (0.68–1.46)
0.97 (0.48–1.26)
NS
0.98 (0.48–1.46)
1.08 (0.81–1.26)
0.87 (0.63–1.29)
0.01
MOS
8q11
39.7%
1.20 (0.78–2.02)
1.31 (0.73–1.96)
NS
1.34 (0.92–2.02)
1.15 (0.77–1.71)
1.23 (0.73–1.96)
NS
EYA1
8q13.3
9.5%
1.08 (0.55–2.07)
0.96 (0.65–1.35)
NS
1.01 (0.66–2.07)
1.05 (0.75–1.64)
0.86 (0.55–1.25)
NS
TPD52
8q21
25.4%
1.26 (0.67–1.98)
1.15 (0.68–1.59)
NS
1.20 (0.90–1.98)
1.22 (0.96–1.74)
1.04 (0.67–1.59)
NS
RAD54B
8q21.3
34.9%
1.33 (0.88–2.29)
1.22 (0.90–2.06)
NS
1.30 (0.90–2.29)
1.20 (0.88–2.06)
1.19 (0.90–1.59)
NS
EIF3S6
8q22
28.6%
1.27 (0.85–1.82)
1.12 (0.64–1.74)
0.04
1.19 (0.74–2.82)
1.13 (0.64–1.70)
1.18 (0.76–1.59)
NS
MYC
8q24.12
73.0%
1.75 (0.95–3.22)
1.76 (0.90–5.81)
NS
1.74 (0.94–3.22)
1.74 (0.90–5.81)
1.84 (1.03–2.74)
NS
WISP1
8q24
36.5%
1.26 (0.79–1.68)
1.26 (0.51–2.08)
NS
1.28 (0.51–1.88)
1.20 (0.79–1.64)
1.27 (0.79–2.08)
NS
KCNK9
8q24.3
34.9%
1.16 (0.59–2.92)
1.24 (0.58–1.90)
NS
1.22 (0.77–1.90
1.11 (0.59–1.87)
1.37 (0.58–2.92)
NS
PTK2
8q24
47.6%
1.40 (0.96–3.47)
1.29 (0.83–1.97)
NS
1.33 (0.83–1.86)
1.25 (0.91–1.97)
1.51 (0.63–5.02)
NS
PTP4A3
8q24.3
33.3%
1.37 (0.62–5.02)
1.23 (0.63–2.24)
NS
1.27 (0.86–2.20)
1.18 (0.62–2.24)
1.55 (0.63–5.02)
NS
PSPC1
13q11
22.2%
1.12 (0.65–1.48)
1.18 (0.71–1.75)
NS
1.20 (0.89–1.75)
1.15 (0.82–1.48)
1.01 (0.65–1.51)
NS
ZNF198
13q11
20.6%
1.05 (0.38–2.17)
1.08 (0.55–2.04)
NS
1.08 (0.55–2.04)
1.15 (0.74–2.17)
0.88 (0.38–1.41)
NS
RASL11A
13q12.2
11.1%
1.09 (0.87–1.35)
1.10 (0.61–1.66)
NS
1.15 (0.84–1.66)
1.08 (0.75–1.52)
0.93 (0.61–1.15)
0.03
CDX2
13q12
22.2%
1.09 (0.70–2.11)
1.19 (0.67–2.09)
NS
1.21 (0.76–2.11)
1.14 (0.70–1.73)
0.91 (0.67–1.30)
0.03
FLT3
13q12
12.7%
1.04 (0.56–2.23)
1.05 (0.65–1.70)
NS
1.08 (0.69–2.23)
1.05 (0.71–1.55)
0.87 (0.56–1.51)
NS
FLT1
13q12.2
20.6%
1.10 (0.85–1.42)
1.13 (0.67–1.85)
NS
1.17 (0.77–1.85)
1.09 (0.82–1.48)
0.98 (0.67–1.34)
NS
BRCA2
13q12.3
22.2%
1.15 (0.53–1.85)
1.22 (0.91–2.62)
NS
1.23 (0.83–2.62)
1.17 (0.93–1.85)
1.08 (0.53–1.31)
NS
FOXO1A
13q14.1
28.6%
1.18 (0.80–1.58)
1.19 (0.79–1.59)
NS
1.20 (0.79–1.59)
1.15 (0.80–1.45)
1.18 (0.89–1.46)
NS
EPSTI1
13q14.11
17.5%
1.51 (1.02–6.32)
1.21 (0.62–2.20)
NS
1.30 (0.94–2.20)
1.17 (0.62–1.68)
1.70 (0.90–6.32)
NS
RB1
13q14.2
14.3%
1.12 (0.72–1.87)
1.04 (0.60–1.97)
NS
1.06 (0.69–1.89)
1.13 (0.60–1.97
1.01 (0.72–1.55)
NS
ATP7B
13q14.2
28.6%
1.15 (0.77–1.45)
1.21 (0.90–1.99)
NS
1.24 (0.91–1.99)
1.14 (0.86–1.53)
1.05 (0.77–1.42)
NS
DACH
13q21.32
20.6%
1.13 (0.82–1.42)
1.13 (0.62–1.72)
NS
1.15 (0.62–1.72)
1.15 (0.88–1.42)
1.05 (0.71–1.36)
NS
ZCCHC3
20p13–p12.2
38.1%
1.19 (0.81–1.82)
1.28 (0.94–2.29)
NS
1.26 (0.81–2.29)
1.21 (0.91–1.92)
1.27 (0.94–1.96)
NS
PCNA
20pter–p12
17.5%
1.09 (0.48–1.65)
1.01 (0.45–1.61)
NS
1.02 (0.45–1.65)
1.06 (0.64–1.50)
1.06 (0.48–1.61)
NS
JAG1
20p12.1–p11.23
42.9%
1.24 (0.69–2.05)
1.29 (0.85–2.13)
NS
1.31 (0.69–2.13)
1.17 (0.85–1.68)
1.31 (0.97–2.05)
NS
THBD
20p12–cen
31.7%
1.04 (0.48–1.79)
1.26 (0.71–3.81)
NS
1.25 (0.48–3.81)
1.07 (0.60–1.66)
1.10 (0.74–1.34)
NS
KIAA0980
20p11.22–p11.1
42.9%
1.17 (0.70–1.76)
1.31 (0.84–2.73)
NS
1.32 (0.70–2.73)
1.12 (0.77–1.55)
1.29 (0.92–1.76)
NS
REM1
20q11.21
34.9%
1.19 (0.42–2.43)
1.24 (0.75–2.21)
NS
1.29 (0.42–2.43)
1.08 (0.60–1.51)
1.24 (0.75–1.79)
NS
BCL2L1
20q11.21
73.0%
1.44 (0.97–1.98)
1.45 (1.04–2.44)
NS
1.50 (1.10–2.44)
1.31 (0.97–1.76)
1.51 (1.19–1.98)
NS
ID1
20q11
29.2%
1.29 (0.66–2.25)
1.41 (0.80–3.33)
NS
1.48 (0.77–3.33)
1.15 (0.66–1.62)
1.32 (1.00–1.73)
NS
TPX2
20q11.2
27.0%
1.26 (0.58–2.22)
1.21 (0.71–2.35)
NS
1.28 (0.58–2.35)
1.13 (0.80–1.57)
1.21 (0.97–1.75)
NS
HCK
20q11–q12
46.0%
1.23 (0.71–2.27)
1.37 (0.90–3.12)
NS
1.40 (0.71–3.12)
1.16 (0.73–1.58)
1.28 (0.93–1.69)
NS
DNMT3B
20q11.2
54.0%
1.29 (0.43–2.26)
1.42 (0.72–2.60)
NS
1.42 (0.43–2.55)
1.20 (0.54–1.74)
1.51 (0.94–2.60)
NS
MAPRE1
20q11.21
20.6%
1.16 (0.84–1.78)
1.14 (0.70–1.78)
NS
1.15 (0.70–1.78)
1.12 (0.84–1.53)
1.17 (0.83–1.54)
NS
E2F1
20q11.2
44.4%
1.35 (0.80–2.31)
1.33 (0.93–2.26)
NS
1.37 (0.82–2.26)
1.21 (0.80–1.72)
1.47 (0.95–2.31)
NS
SRC
20q12–q13
57.1%
1.38 (0.71–2.12)
1.45 (0.77–2.70)
NS
1.51 (0.88–2.70)
1.22 (0.71–1.67)
1.48 (1.02–2.12)
NS
NNAT
20q11.2–q12
52.4%
1.30 (0.47–2.50)
1.30 (0.55–2.65)
NS
1.36 (0.47–2.65)
1.16 (0.69–1.78)
1.27 (0.91–1.91)
NS
TOP1
20q12–q13.1
63.5%
1.57 (0.85–2.78)
1.47 (0.88–2.40)
NS
1.57 (1.00–2.78)
1.29 (0.85–1.72)
1.67 (0.94–2.69)
NS
MYBL2
20q13.1
73.0%
1.49 (0.58–2.79)
1.73 (0.70–3.83)
NS
1.79 (0.58–3.83)
1.34 (0.58–2.51)
1.62 (0.95–2.65)
NS
ADA
20q12–q13.11
41.3%
1.17 (0.42–1.80)
1.28 (0.67–2.24)
NS
1.32 (0.42–2.24)
1.08 (0.53–1.74)
1.22 (0.74–1.80)
NS
WISP2
20q12–q13.1
52.4%
1.31 (0.78–2.03)
1.33 (0.78–2.62)
NS
1.37 (0.82–2.62)
1.18 (0.78–1.69)
1.40 (0.95–2.03)
NS
MMP9
20q11.2–q13.1
47.6%
1.26 (0.76–2.06)
1.33 (0.87–2.37)
NS
1.38 (0.81–2.37)
1.18 (0.76–1.72)
1.27 (0.87–1.81)
NS
CSE1L
20q13
49.2%
1.40 (0.85–2.65)
1.31 (0.88–2.54)
NS
1.37 (0.85–2.65)
1.25 (0.94–1.60)
1.43 (1.11–1.82)
NS
PTPN1
20q13.1–q13.2
65.1%
1.46 (0.85–2.76)
1.51 (0.87–2.62)
NS
1.55 (0.85–2.76)
1.32 (0.96–1.82)
1.57 (1.04–2.54)
NS
NFATC2
20q13.2–q13.3
55.6%
1.31 (0.63–2.34)
1.60 (0.77–5.60)
NS
1.61 (0.63–5.60)
1.27 (0.65–2.09)
1.45 (0.83–2.50)
NS
ZNF217
20q13.2
57.1%
1.74 (0.99–6.65)
1.79 (0.96–10.09)
NS
2.06 (1.05–10.09)
1.27 (0.96–1.82)
1.50 (1.15–2.09)
0.02
BCAS1
20q13.2–q13.3
58.7%
1.42 (0.80–2.77)
1.94 (0.95–12.17)
NS
2.03 (0.80–12.17)
1.29 (0.85–1.88)
1.47 (1.05–2.19)
NS
CYP24A1
20q13
60.3%
1.47 (0.85–3.43)
1.81 (0.80–9.16)
NS
1.92 (0.85–9.16)
1.26 (0.80–1.91)
1.52 (0.96–2.27)
NS
STX16
20q13.32
77.8%
1.78 (0.91–5.05)
1.81 (0.95–3.95)
NS
1.90 (1.11–3.95)
1.47 (0.91–2.62)
2.03 (1.09–5.05)
0.03
GNAS
20q13.3
76.2%
2.00 (0.94–7.97)
1.82 (0.87–4.68)
NS
2.02 (0.94–7.97)
1.48 (0.87–2.26)
2.12 (1.16–4.99)
NS
EEF1A2
20q13.3
47.6%
1.20 (0.41–2.52)
1.41 (0.63–2.20)
NS
1.39 (0.41–2.52)
1.21 (0.42–2.18)
1.40 (0.83–2.09)
NS
TNFRSF6B
20q13.3
49.2%
1.20 (0.58–2.51)
1.45 (0.82–3.21)
0.02
1.41 (0.67–3.21)
1.25 (0.58–2.01)
1.36 (0.82–2.13)
NS
KCNQ2
20q13.3
38.1%
1.18 (0.49–2.59)
1.30 (0.43–2.08)
NS
1.27 (0.43–2.08)
1.13 (0.52–1.85)
1.48 (0.55–2.59)
NS
GATA5
20q13.33
60.3%
1.44 (0.60–4.15)
1.58 (0.62–4.12)
NS
1.54 (0.63–4.12)
1.38 (0.60–2.46)
1.82 (0.62–4.15)
NS
FLJ20517
20q13.33
41.3%
1.28 (0.65–3.54)
1.34 (0.80–2.38)
NS
1.30 (0.80–2.38)
1.24 (0.65–1.74)
1.60 (0.95–3.54)
NS
UCKL1
20q13.33
39.7%
1.13 (0.56–2.03)
1.30 (0.67–2.86)
NS
1.29 (0.56–2.86)
1.13 (0.67–1.87)
1.24 (0.85–1.65)
NS
OPRL1
20q13.33
28.6%
1.15 (0.49–2.70)
1.19 (0.62–2.28)
NS
1.18 (0.49–2.28)
1.08 (0.51–1.57)
1.38 (0.67–2.70)
NS

Correlation of copy-number status of MLPA and array CGH

The BAC clones RP4-583P15 and RP4-724E16 comprised the location of the TNFRSF6B and ZNF217 genes, respectively. To evaluate the correlation between the array CGH and MLPA data, the MLPA DNA copy-number ratios were transformed to a log2 scale. Spearman correlation yielded a significant correlation between array CGH and MLPA data for both TNFRSF6B (r = 0.57, p < 0.001; Fig. 4a) and ZNF217 (r = 0.51, p < 0.001; Fig. 4b).

Discussion

Gastric cancer is a common disease with a poor prognosis [1]. Chromosomal instability is a major mechanism of genetic instability in gastric cancers which has been widely studied by array CGH analysis [2, 32]. Frequent copy-number gains of chromosomes 8q, 13q, and 20q have been detected in gastric adenocarcinomas by this technique [310]. In this study, we report a detailed analysis of DNA copy-number changes of genes located on chromosomes 8, 13, and 20 in gastric adenocarcinomas aiming to correlate gene-specific alterations to clinicopathological data.
In the present study of 63 gastric adenocarcinomas studied by array CGH, nearly 80% of the cancers showed gain of chromosomes 8q and 20q. These frequencies of gains are consistent with previous studies where gains of chromosomes 8q and 20q were reported in 40–80% and 50–80% of gastric cancers, respectively [4, 10, 13, 14, 17, 33]. Both gains and losses of chromosome 13q have been detected in gastric cancers. Losses of chromosome 13q have been described in up to 30% of gastric cancers and gains of 13q have been described in up to 40% of gastric cancers [4, 10, 1315, 17, 33], which is consistent with the findings of the present study.
When analyzing DNA copy-number gain at the gene level, mean copy-number ratio of two genes, EIF3S6E (8q22) and TNFRSF6B (20q12.3), correlated significantly with lymph node status. Mean copy-number ratios of EIF3S6E were within the normal range for both lymph node-positive and lymph node-negative gastric cancers, corresponding to the array CGH results in this study. Mean copy-number ratio of TNFRSF6B was significantly higher in lymph node-positive compared with lymph node-negative gastric cancers and was within the range of DNA copy-number gain and normal, respectively. Although 20q gain CGH data correlated with lymph node metastasis, of the 20q genes tested, only DNA copy-number gain of TNFRSF6B was significantly correlated with lymph node metastasis. TNFRSF6B is a member of the tumor necrosis factor receptor superfamily and is also known as decoy receptor 3 (DrR3). TNFRSF6B binds to FasL and inhibits FasL-induced apoptosis [34]. Overexpression of this gene has been observed in multiple cancer types, including cancers of the gastrointestinal tract. Interestingly, gastric cancers with pN2 and pN3 diseases have been shown to have significantly higher TNFRSF6B expression compared with gastric cancers with pN0 and pN1 diseases. Serum levels of TNFRSF6B were correlated with TNM stage in gastric cancers [35, 36]. We found TNFRSF6B DNA copy-number ratios on average to be gained in gastric cancers metastasized to the lymph nodes (pN1–3) compared with on average normal DNA copy-number ratios of TNFRSF6B in non-metastasized gastric cancers (pN0). Although previous studies have reported overexpression of TNFRSF6B without gene amplification [37], results of the present study indicate that DNA copy-number status of TNFRSF6B could be a valuable marker for identifying lymph node-positive gastric cancers.
DNA copy-number status of five genes, FBXO25 (8p23.3), RASL11A (13q12.2), CDX2 (13q12), ZNF217 (20q13.2), and STX16 (20q13.32), correlated significantly with histological tumor type, i.e., intestinal-, diffuse-, or mixed-type gastric cancer. Mean DNA copy-number ratios of FBXO25, RASL11A, and CDX2, however, were within the normal copy-number range for all three histological types, in consistence with the array CGH data of this study. The biological meaning of this difference, therefore, is not immediately clear, but could, e.g., relate to only a subpopulation of tumor cells being affected.
STX16 showed significantly higher mean copy-number ratios in intestinal- and mixed-type compared with diffuse-type gastric cancers; however, DNA copy-number gain was observed in all three tumor groups. STX16 encodes a syntaxin protein which plays a role in intracellular trafficking [38], but its role in cancer still has to be elucidated.
ZNF217 encodes a transcription factor which was shown to be involved in immortalization of breast cancer cells when overexpressed [39]. High-level amplifications of ZNF217 have been previously described in approximately 10% of gastric cancers [9, 10, 40]. In the study of Weiss et al. on three tumors, all of the intestinal-type, out of 27 gastric adenocarcinomas, showed an amplification on 20q13. Fluorescence in situ hybridization (FISH) analysis was performed on these three tumors, confirming the copy-number increase at this locus [9]. Results from the present study suggest that DNA copy-number gain of ZNF217 plays a more important role in intestinal- and mixed-type compared with diffuse-type gastric cancers, since gains of mean copy-number ratios are observed in intestinal- and mixed-type gastric cancers and normal mean copy-number ratio in diffuse-type gastric cancers.
In summary, we present a detailed DNA copy-number analysis of a panel of genes located on chromosomes 8, 13, and 20 in gastric adenocarcinomas. We found that DNA copy-number gain of ZNF217 plays a role mainly in intestinal- and mixed-type but not in diffuse-type gastric adenocarcinomas. In addition, we showed that DNA copy-number gain of TNFRSF6B is significantly correlated to gastric cancers with lymph node metastasis, indicating a potential role for this gene as a biomarker for identifying patients at high risk of lymph node metastasis, who might, therefore, benefit from extended lymph node resection or neoadjuvant chemotherapy to improve gastric cancer outcome.

Acknowledgements

This study was financially supported by the Dutch Cancer Society, grant KWF2004-3051.

Conflict of interest statement

We declare that we have no conflict of interest.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.
Open AccessThis is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License (https://​creativecommons.​org/​licenses/​by-nc/​2.​0), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Parkin DM, Bray F, Ferlay J et al (2005) Global cancer statistics, 2002. CA Cancer J Clin 55:74–108PubMedCrossRef Parkin DM, Bray F, Ferlay J et al (2005) Global cancer statistics, 2002. CA Cancer J Clin 55:74–108PubMedCrossRef
2.
Zurück zum Zitat Lengauer C, Kinzler KW, Vogelstein B (1998) Genetic instabilities in human cancers. Nature 396:643–649PubMedCrossRef Lengauer C, Kinzler KW, Vogelstein B (1998) Genetic instabilities in human cancers. Nature 396:643–649PubMedCrossRef
3.
Zurück zum Zitat Hidaka S, Yasutake T, Kondo M et al (2003) Frequent gains of 20q and losses of 18q are associated with lymph node metastasis in intestinal-type gastric cancer. Anticancer Res 23:3353–3357PubMed Hidaka S, Yasutake T, Kondo M et al (2003) Frequent gains of 20q and losses of 18q are associated with lymph node metastasis in intestinal-type gastric cancer. Anticancer Res 23:3353–3357PubMed
4.
Zurück zum Zitat Kimura Y, Noguchi T, Kawahara K et al (2004) Genetic alterations in 102 primary gastric cancers by comparative genomic hybridization: gain of 20q and loss of 18q are associated with tumor progression. Mod Pathol 17:1328–1337PubMedCrossRef Kimura Y, Noguchi T, Kawahara K et al (2004) Genetic alterations in 102 primary gastric cancers by comparative genomic hybridization: gain of 20q and loss of 18q are associated with tumor progression. Mod Pathol 17:1328–1337PubMedCrossRef
5.
Zurück zum Zitat Koo SH, Kwon KC, Shin SY et al (2000) Genetic alterations of gastric cancer: comparative genomic hybridization and fluorescence In situ hybridization studies. Cancer Genet Cytogenet 117:97–103PubMedCrossRef Koo SH, Kwon KC, Shin SY et al (2000) Genetic alterations of gastric cancer: comparative genomic hybridization and fluorescence In situ hybridization studies. Cancer Genet Cytogenet 117:97–103PubMedCrossRef
6.
Zurück zum Zitat Sakakura C, Mori T, Sakabe T et al (1999) Gains, losses, and amplifications of genomic materials in primary gastric cancers analyzed by comparative genomic hybridization. Genes Chromosomes Cancer 24:299–305PubMedCrossRef Sakakura C, Mori T, Sakabe T et al (1999) Gains, losses, and amplifications of genomic materials in primary gastric cancers analyzed by comparative genomic hybridization. Genes Chromosomes Cancer 24:299–305PubMedCrossRef
7.
Zurück zum Zitat Takada H, Imoto I, Tsuda H et al (2005) Screening of DNA copy-number aberrations in gastric cancer cell lines by array-based comparative genomic hybridization. Cancer Sci 96:100–110PubMedCrossRef Takada H, Imoto I, Tsuda H et al (2005) Screening of DNA copy-number aberrations in gastric cancer cell lines by array-based comparative genomic hybridization. Cancer Sci 96:100–110PubMedCrossRef
8.
Zurück zum Zitat van Grieken NC, Weiss MM, Meijer GA et al (2000) Helicobacter pylori-related and -non-related gastric cancers do not differ with respect to chromosomal aberrations. J Pathol 192:301–306PubMedCrossRef van Grieken NC, Weiss MM, Meijer GA et al (2000) Helicobacter pylori-related and -non-related gastric cancers do not differ with respect to chromosomal aberrations. J Pathol 192:301–306PubMedCrossRef
9.
Zurück zum Zitat Weiss MM, Snijders AM, Kuipers EJ et al (2003) Determination of amplicon boundaries at 20q13.2 in tissue samples of human gastric adenocarcinomas by high-resolution microarray comparative genomic hybridization. J Pathol 200:320–326PubMedCrossRef Weiss MM, Snijders AM, Kuipers EJ et al (2003) Determination of amplicon boundaries at 20q13.2 in tissue samples of human gastric adenocarcinomas by high-resolution microarray comparative genomic hybridization. J Pathol 200:320–326PubMedCrossRef
10.
Zurück zum Zitat Weiss MM, Kuipers EJ, Postma C et al (2004) Genomic alterations in primary gastric adenocarcinomas correlate with clinicopathological characteristics and survival. Cell Oncol 26:307–317PubMed Weiss MM, Kuipers EJ, Postma C et al (2004) Genomic alterations in primary gastric adenocarcinomas correlate with clinicopathological characteristics and survival. Cell Oncol 26:307–317PubMed
11.
Zurück zum Zitat Wu MS, Shun CT, Wang HP et al (1997) Genetic alterations in gastric cancer: relation to histological subtypes, tumor stage, and Helicobacter pylori infection. Gastroenterology 112:1457–1465PubMedCrossRef Wu MS, Shun CT, Wang HP et al (1997) Genetic alterations in gastric cancer: relation to histological subtypes, tumor stage, and Helicobacter pylori infection. Gastroenterology 112:1457–1465PubMedCrossRef
12.
Zurück zum Zitat Buffart TE, Carvalho B, Mons T et al (2007) DNA copy-number profiles of gastric cancer precursor lesions. BMC Genomics 8:345PubMedCrossRef Buffart TE, Carvalho B, Mons T et al (2007) DNA copy-number profiles of gastric cancer precursor lesions. BMC Genomics 8:345PubMedCrossRef
13.
Zurück zum Zitat Kim YH, Kim NG, Lim JG et al (2001) Chromosomal alterations in paired gastric adenomas and carcinomas. Am J Pathol 158:655–662PubMed Kim YH, Kim NG, Lim JG et al (2001) Chromosomal alterations in paired gastric adenomas and carcinomas. Am J Pathol 158:655–662PubMed
14.
Zurück zum Zitat Kokkola A, Monni O, Puolakkainen P et al (1998) Presence of high-level DNA copy-number gains in gastric carcinoma and severely dysplastic adenomas but not in moderately dysplastic adenomas. Cancer Genet Cytogenet 107:32–36PubMedCrossRef Kokkola A, Monni O, Puolakkainen P et al (1998) Presence of high-level DNA copy-number gains in gastric carcinoma and severely dysplastic adenomas but not in moderately dysplastic adenomas. Cancer Genet Cytogenet 107:32–36PubMedCrossRef
15.
Zurück zum Zitat van Dekken H, Alers JC, Riegman PH et al (2001) Molecular cytogenetic evaluation of gastric cardia adenocarcinoma and precursor lesions. Am J Pathol 158:1961–1967PubMed van Dekken H, Alers JC, Riegman PH et al (2001) Molecular cytogenetic evaluation of gastric cardia adenocarcinoma and precursor lesions. Am J Pathol 158:1961–1967PubMed
16.
Zurück zum Zitat Weiss MM, Kuipers EJ, Postma C et al (2003) Genome wide array comparative genomic hybridisation analysis of premalignant lesions of the stomach. Mol Pathol 56:293–298PubMedCrossRef Weiss MM, Kuipers EJ, Postma C et al (2003) Genome wide array comparative genomic hybridisation analysis of premalignant lesions of the stomach. Mol Pathol 56:293–298PubMedCrossRef
17.
Zurück zum Zitat Buffart T, Carvalho B, Hopmans E et al (2007) Gastric cancers in young and elderly patients show different genomic profiles. J Pathol 211:45–51PubMedCrossRef Buffart T, Carvalho B, Hopmans E et al (2007) Gastric cancers in young and elderly patients show different genomic profiles. J Pathol 211:45–51PubMedCrossRef
18.
Zurück zum Zitat Weiss MM, Kuipers EJ, Postma C et al (2003) Genomic profiling of gastric cancer predicts lymph node status and survival. Oncogene 22:1872–1879PubMedCrossRef Weiss MM, Kuipers EJ, Postma C et al (2003) Genomic profiling of gastric cancer predicts lymph node status and survival. Oncogene 22:1872–1879PubMedCrossRef
19.
Zurück zum Zitat Carvalho B, Postma C, Mongera S et al (2008) Integration of DNA and expression microarray data unravels seven putative oncogenes on 20Q amplicon involved in colorectal adenoma to carcinoma progression. Cell Oncol 30:145–146 Carvalho B, Postma C, Mongera S et al (2008) Integration of DNA and expression microarray data unravels seven putative oncogenes on 20Q amplicon involved in colorectal adenoma to carcinoma progression. Cell Oncol 30:145–146
20.
Zurück zum Zitat Hermsen M, Postma C, Baak J et al (2002) Colorectal adenoma to carcinoma progression follows multiple pathways of chromosomal instability. Gastroenterology 123:1109–1119PubMedCrossRef Hermsen M, Postma C, Baak J et al (2002) Colorectal adenoma to carcinoma progression follows multiple pathways of chromosomal instability. Gastroenterology 123:1109–1119PubMedCrossRef
21.
Zurück zum Zitat Aust DE, Muders M, Kohler A et al (2004) Prognostic relevance of 20q13 gains in sporadic colorectal cancers: a FISH analysis. Scand J Gastroenterol 39:766–772PubMedCrossRef Aust DE, Muders M, Kohler A et al (2004) Prognostic relevance of 20q13 gains in sporadic colorectal cancers: a FISH analysis. Scand J Gastroenterol 39:766–772PubMedCrossRef
22.
Zurück zum Zitat Postma C, Terwischa S, Hermsen MA et al (2007) Gain of chromosome 20q is an indicator of poor prognosis in colorectal cancer. Cell Oncol 29:73–75PubMed Postma C, Terwischa S, Hermsen MA et al (2007) Gain of chromosome 20q is an indicator of poor prognosis in colorectal cancer. Cell Oncol 29:73–75PubMed
23.
Zurück zum Zitat Schouten JP, McElgunn CJ, Waaijer R et al (2002) Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe amplification. Nucleic Acids Res 30:e57PubMedCrossRef Schouten JP, McElgunn CJ, Waaijer R et al (2002) Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe amplification. Nucleic Acids Res 30:e57PubMedCrossRef
24.
Zurück zum Zitat Buffart TE, Coffa J, Hermsen MA et al (2005) DNA copy-number changes at 8q11–24 in metastasized colorectal cancer. Cell Oncol 27:57–65PubMed Buffart TE, Coffa J, Hermsen MA et al (2005) DNA copy-number changes at 8q11–24 in metastasized colorectal cancer. Cell Oncol 27:57–65PubMed
25.
Zurück zum Zitat Postma C, Hermsen MA, Coffa J et al (2005) Chromosomal instability in flat adenomas and carcinomas of the colon. J Pathol 205:514–521PubMedCrossRef Postma C, Hermsen MA, Coffa J et al (2005) Chromosomal instability in flat adenomas and carcinomas of the colon. J Pathol 205:514–521PubMedCrossRef
26.
Zurück zum Zitat Bonenkamp JJ, Hermans J, Sasako M et al (1999) Extended lymph-node dissection for gastric cancer. Dutch Gastric Cancer Group. N Engl J Med 340:908–914PubMedCrossRef Bonenkamp JJ, Hermans J, Sasako M et al (1999) Extended lymph-node dissection for gastric cancer. Dutch Gastric Cancer Group. N Engl J Med 340:908–914PubMedCrossRef
27.
Zurück zum Zitat Buffart TE, Tijssen M, Krugers T et al (2007) DNA quality assessment for array CGH by isothermal whole genome amplification. Cell Oncol 29:351–359PubMed Buffart TE, Tijssen M, Krugers T et al (2007) DNA quality assessment for array CGH by isothermal whole genome amplification. Cell Oncol 29:351–359PubMed
28.
Zurück zum Zitat Snijders AM, Nowak N, Segraves R et al (2001) Assembly of microarrays for genome-wide measurement of DNA copy-number. Nat Genet 29:263–264PubMedCrossRef Snijders AM, Nowak N, Segraves R et al (2001) Assembly of microarrays for genome-wide measurement of DNA copy-number. Nat Genet 29:263–264PubMedCrossRef
29.
Zurück zum Zitat van de Wiel MA, Kim KI, Vosse SJ et al (2007) CGHcall: calling aberrations for array CGH tumor profiles. Bioinformatics 23:892–894PubMedCrossRef van de Wiel MA, Kim KI, Vosse SJ et al (2007) CGHcall: calling aberrations for array CGH tumor profiles. Bioinformatics 23:892–894PubMedCrossRef
30.
Zurück zum Zitat Coffa J, van de Wiel MA, Diosdado B et al (2008) MLPAnalyzer: data analysis tool for reliable automated normalization of MLPA fragment data. Cell Oncol 30:323–335PubMed Coffa J, van de Wiel MA, Diosdado B et al (2008) MLPAnalyzer: data analysis tool for reliable automated normalization of MLPA fragment data. Cell Oncol 30:323–335PubMed
31.
Zurück zum Zitat Laurén P (1965) The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attemt at a histo-clinical classification. Acta Pathol Microbiol Scand 64:31–49PubMed Laurén P (1965) The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attemt at a histo-clinical classification. Acta Pathol Microbiol Scand 64:31–49PubMed
32.
Zurück zum Zitat Pinkel D, Albertson DG (2005) Array comparative genomic hybridization and its applications in cancer. Nat Genet 37(Suppl):S11–S17PubMedCrossRef Pinkel D, Albertson DG (2005) Array comparative genomic hybridization and its applications in cancer. Nat Genet 37(Suppl):S11–S17PubMedCrossRef
33.
Zurück zum Zitat Carvalho B, Buffart TE, Reis RM et al (2006) Mixed gastric carcinomas show similar chromosomal aberrations in both their diffuse and glandular components. Cell Oncol 28:283–294PubMed Carvalho B, Buffart TE, Reis RM et al (2006) Mixed gastric carcinomas show similar chromosomal aberrations in both their diffuse and glandular components. Cell Oncol 28:283–294PubMed
34.
Zurück zum Zitat Pitti RM, Marsters SA, Lawrence DA et al (1998) Genomic amplification of a decoy receptor for Fas ligand in lung and colon cancer. Nature 396:699–703PubMedCrossRef Pitti RM, Marsters SA, Lawrence DA et al (1998) Genomic amplification of a decoy receptor for Fas ligand in lung and colon cancer. Nature 396:699–703PubMedCrossRef
35.
Zurück zum Zitat Wu Y, Han B, Sheng H et al (2003) Clinical significance of detecting elevated serum DcR3/TR6/M68 in malignant tumor patients. Int J Cancer 105:724–732PubMedCrossRef Wu Y, Han B, Sheng H et al (2003) Clinical significance of detecting elevated serum DcR3/TR6/M68 in malignant tumor patients. Int J Cancer 105:724–732PubMedCrossRef
36.
Zurück zum Zitat Wu Y, Guo E, Yu J et al (2008) High DcR3 expression predicts stage pN2–3 in gastric cancer. Am J Clin Oncol 31:79–83PubMedCrossRef Wu Y, Guo E, Yu J et al (2008) High DcR3 expression predicts stage pN2–3 in gastric cancer. Am J Clin Oncol 31:79–83PubMedCrossRef
37.
Zurück zum Zitat Bai C, Connolly B, Metzker ML et al (2000) Overexpression of M68/DcR3 in human gastrointestinal tract tumors independent of gene amplification and its location in a four-gene cluster. Proc Natl Acad Sci U S A 97:1230–1235PubMedCrossRef Bai C, Connolly B, Metzker ML et al (2000) Overexpression of M68/DcR3 in human gastrointestinal tract tumors independent of gene amplification and its location in a four-gene cluster. Proc Natl Acad Sci U S A 97:1230–1235PubMedCrossRef
38.
Zurück zum Zitat Simonsen A, Bremnes B, Ronning E et al (1998) Syntaxin-16, a putative Golgi t-SNARE. Eur J Cell Biol 75:223–231PubMed Simonsen A, Bremnes B, Ronning E et al (1998) Syntaxin-16, a putative Golgi t-SNARE. Eur J Cell Biol 75:223–231PubMed
39.
Zurück zum Zitat Nonet GH, Stampfer MR, Chin K et al (2001) The ZNF217 gene amplified in breast cancers promotes immortalization of human mammary epithelial cells. Cancer Res 61:1250–1254PubMed Nonet GH, Stampfer MR, Chin K et al (2001) The ZNF217 gene amplified in breast cancers promotes immortalization of human mammary epithelial cells. Cancer Res 61:1250–1254PubMed
40.
Zurück zum Zitat Yang SH, Seo MY, Jeong HJ et al (2005) Gene copy-number change events at chromosome 20 and their association with recurrence in gastric cancer patients. Clin Cancer Res 11:612–620PubMed Yang SH, Seo MY, Jeong HJ et al (2005) Gene copy-number change events at chromosome 20 and their association with recurrence in gastric cancer patients. Clin Cancer Res 11:612–620PubMed
Metadaten
Titel
High resolution analysis of DNA copy-number aberrations of chromosomes 8, 13, and 20 in gastric cancers
verfasst von
Tineke E. Buffart
Nicole C. T. van Grieken
Marianne Tijssen
Jordy Coffa
Bauke Ylstra
Heike I. Grabsch
Cornelis J. H. van de Velde
Beatriz Carvalho
Gerrit A. Meijer
Publikationsdatum
01.09.2009
Verlag
Springer-Verlag
Erschienen in
Virchows Archiv / Ausgabe 3/2009
Print ISSN: 0945-6317
Elektronische ISSN: 1432-2307
DOI
https://doi.org/10.1007/s00428-009-0814-y

Weitere Artikel der Ausgabe 3/2009

Virchows Archiv 3/2009 Zur Ausgabe

Neu im Fachgebiet Pathologie