Analysis of genomic aberrations associated with the clinicopathological parameters of rectal cancer by array‑based comparative genomic hybridization

  • Authors:
    • Jian-Wei Liang
    • Zhi-Zhou Shi
    • Tong-Tong Zhang
    • Jia-Jie Hao
    • Zheng Wang
    • Xiao-Min Wang
    • Hai Yang
    • Ming-Rong Wang
    • Zhi-Xiang Zhou
    • Yu Zhang
  • View Affiliations

  • Published online on: February 21, 2013     https://doi.org/10.3892/or.2013.2296
  • Pages: 1827-1834
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to screen and identify the chromosomal aberrations that are correlated with clinicopathological characteristics of rectal cancer using array-based comparative genomic hybridization (array-CGH). Forty-eight fresh frozen tumor tissues of rectal carcinoma were analyzed by array-CGH. The results showed that most frequent gains included 8q24.3, 20q11.21-q13.32, 20q13.33 and losses in 8p23.3-p12, 17p13.1-p12 and 18q11.2-q23 were noted. Fourteen amplifications and seven homozygous deletions were identified in the rectal cancer samples. Losses of 4p16.1-p15.31, 8p21.1-p12 and gains of 7p12.3-p12.1 and 13q33.1-q34 were associated with positive lymph node status and advanced clinical stage (stages III and IV). The 17q24.2-25.3 gain was more frequent in patients with distant metastasis. Integrated analysis indicated that overexpression of PDP1, TRIB1, C13orf27, FOXA2, PMEPA1 and PHACTR3 was associated with gains, and underexpression of FHOD, SMAD4 and BCL2 was associated with losses. Pathway enrichment analysis showed that pathways of nitrogen metabolism, oxidative phosphorylation, cell cycle, maturity onset diabetes of young, cytokine-cytokine receptor interaction, MAPK signaling pathway and dentatorubropallidoluysian atrophy were influenced by copy number changes.

Introduction

Colorectal cancer (CRC) is a common malignant tumor worldwide, and over 1.2 million new cases and 608,700 deaths were estimated to have occurred in 2008 (1). The incidence of CRC in China has increased rapidly since the 1980s (2,3). Currently, CRC is the fifth leading cause of cancer-related deaths (4).

CRC can be divided into two types based on genetic abnormalities (5,6). The major type is the chromosomal instability phenotype, which consists of more than 85% of CRCs and is characterized by frequent chromosomal imbalances. The minor type is the microsatellite instability phenotype, which exhibits microsatellite instability owing to DNA replication errors and comprises <15% of CRCs. The genomic instability of the two types can lead to DNA copy number aberrations.

Cancers occur as a result of the accumulation of genetic alterations that are associated with carcinogenesis (5,7). Thus, the study of the cancer genome by high-resolution and high-throughput technology, for example array-based comparative genomic hybridization (array-CGH), not only has the ability to clarify the relationship between genomic abnormalities and clinicopathological factors, but may also optimize the treatment of patients by using their cancer genome information. Although several DNA copy number aberrations have been reported to have linkage with clinicopathological characteristics of patients with CRC (810), the available information is still limited particularly in Chinese patients.

The present study identified frequent DNA copy number changes in rectal cancer samples from Chinese patients and candidate target genes using integrated analysis of the genome and gene expression data of NCI-60 cell lines. We evaluated the genetic changes associated with lymph node metastasis, tumor stage and distant metastasis using the methods of frequency plot comparison together with statistical analysis.

Materials and methods

Study design

This study was conducted to identify genomic changes associated with clinicopathological factors and candidate targets of most frequent gains and losses of rectal cancer. We determined the genetic aberrations in 48 rectal carcinomas using Agilent 60K Human Genome CGH microarray and screened those linked with clinicopathological characteristics. We then compared the gene expression profiling of CRC cell lines with or without gains of 8q, 13q, 17q, 20p, 20q or losses of 8p, 11q, 18q and identified genes whose expression was linked with DNA copy number changes.

Patients and samples

Fresh tissues from 48 rectal carcinoma patients were collected at the Department of Pathology, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China. None of the patients received either irradiation or chemotherapy prior to surgery. All of the samples used in this study were residual specimens after diagnostic sampling. Every patient signed a separate informed consent form for sampling and research. Ethics approval was obtained from the Ethics Committee of Cancer Institute and Hospital, Chinese Academy of Medical Sciences. Representative tumor regions were excised by experienced pathologists. The clinicopathologic characteristics of the patients are summarized in Table I.

Table I

Clinicopathological characteristics of the rectal cancer patients in the array-CGH study.

Table I

Clinicopathological characteristics of the rectal cancer patients in the array-CGH study.

Patient no.GenderAge (years)TNMpStage
1M67T3N1M0III B
2M58T3N1M0III B
3M43T4N2M1III C
4F63T3N2M0III C
5F69T4N2M0III C
6F52T3N2M0III C
7M56T3N2M1III C
8F47T4N2M0III C
9F61T4N2M0III C
10M41T3N2M1III C
11M58T3N2M1IV
12F62T3N1M1IV
13F36T3N0M1IV
14M59T3N0M1IV
15M49T3N1M1IV
16M53T3N2M1IV
17M75T3N2M1IV
18F79T3N2M0III C
19F22T4N2M0III C
20F70T4N2M0III C
21M77T4N2M0III C
22M36T3N1M0III B
23M49T3N0M0II A
24M45T4N0M0II B
25M61T3N0M0II A
26M51T3N0M0II A
27F66T3N0M1II A
28M59T3N0M1II A
29M45T3N0M0II A
30M72T3N0M0II A
31M63T3N0M0II A
32M58T3N0M0II A
33M68T3N0M0II A
34M63T3N0M0II A
35M63T3N0M0II A
36M55T3N0M0II A
37M42T3N0M0II A
38F56T3N0M0II A
39F52T4N0M0II B
40M45T4N0M0II B
41M47T4N0M0II B
42M62T4N0M0II B
43M64T3N1M0III B
44F53T4N1M0III B
45F59T3N1M0III B
46F49T3N1M0III B
47F39T3N1M0III B
48M63T3N1M1III B
Genomic DNA extraction

Genomic DNA was isolated from tumor tissues using the Qiagen DNeasy Blood and Tissue kit as described by the manufacturer (Qiagen, Hilden, Germany). Tumor cell content of all the samples was >50% as determined by H&E staining.

Array-based CGH

Array-CGH experiments were performed using standard Agilent protocols (Agilent Technologies, Santa Clara, CA, USA). Commercial human genomic DNA (Promega, Warrington, UK) was used as reference. For each CGH hybridization, 400 ng of reference genomic DNA and the same amount of tumor DNA were digested with AluI and RSAI restriction enzymes (Promega). The digested reference DNA fragments were labeled with cyanine 3-dUTP and the tumor DNA with cyanine 5-dUTP (Agilent Technologies). After clean-up, reference and tumor DNA probes were mixed and hybridized onto an Agilent 60K human genome CGH microarray (Agilent Technologies) for 24 h. Washing, scanning and data extraction procedures were performed following standard protocols.

Microarray data analysis

Microarray data were analyzed using Agilent Genomic Workbench (Agilent Technologies), CGH ArrayTools (http://linus.nci.nih.gov/BRB-ArrayTools.html) and MD-SeeGH (www.flintbox.ca). Agilent Genomic Workbench was used to calculate the log2ratio for every probe and identified genomic aberrations. Mean Log2ratio of all probes in a chromosome region between 0.25 and 0.75 was classified as a genomic gain, >0.75 as high-level DNA amplification, <−0.25 as a hemizygous loss, and <−0.75 as a homozygous deletion. Pathway enrichment analyses were performed by CGH ArrayTools.

Integration analysis of DNA copy number and gene expression data of the NCI 60 cell lines

The DNA copy number and gene expression data of the NCI 60 cell lines were obtained from CellMiner (http://discover.nci.nih.gov/cellminer). We selected the data sets of aCGH Agilent 44K and Agilent mRNA for analysis. The genetic changes of seven CRC cell lines (including colo205, HCT_116, HCT_15, KM12, HCC_2998, HT29 and SW_620) were analyzed and divided into the gain/loss group and no change group. The differentially expressed genes between the two groups were indentified with a cutoff of a 2-fold change using GeneSpring GX (Agilent Technologies).

Oncomine data analysis

The mRNA expression of genes which had a >5-fold change in expression between the gain/loss CRC cell lines and the no change cell lines was analyzed using Oncomine database (https://www.oncomine.org/resource/login.html). Details of standardized normalization techniques and statistical methods can be found on the the Oncomine website. The data of the interested genes in different types of cancer were collected and then their expression status in CRC was analyzed.

Statistical analysis

The Chi-square test was used to analyze the significance of correlation between genomic aberrations and clinical factors. Differences were considered significant at P<0.05.

Results

Recurrent copy number alterations in rectal carcinoma detected by array-CGH

Forty eight samples of rectal carcinoma were analyzed in this study and all of them had genomic changes. The most frequent genomic aberrations were gains of 8q24.3, 20q11.21-q13.32, 20q13.33, and losses of 8p23.3-p12, 17p13.1-p12 and 18q11.2-q23 (Fig. 1A). High-level amplifications were detected at 14 chromosome regions including 7p22.3-p21.3, 7q22.1, 8p11.21, 8p11.23, 8q22.1, 8q24.3, 13q12.2, 13q14.2-q14.3, 13q31.3, 16p11.2, 19p13.2, 20p11.23, 20q11.21-q11.23 and 20q13.33. Homozygous deletions were identified in 5q14.3, 8p23.3-p21.2, 15q11.2, 16p13.2, 17p13.1-p12, 18q21.2 and 20p12.1 (Table II). After analyzing the number of changes in rectal cancer, we found that nearly half of cases in the array-CGH study had 10 to 29 genetic alterations (Fig. 1B). GISTIC analysis showed that a copy number increase of MTUS2 (13q12.3) and decrease of C3orf57 (3q26.1), SPRYD5 (11q11), OR5W2 (11q11) and MKL1 (22q13.2) were significant in the rectal cancer cases (Fig. 1C).

Table II

High-level amplifications and homozygous deletions in the rectal cancer cases.

Table II

High-level amplifications and homozygous deletions in the rectal cancer cases.

ChangesCytobandStartEndNo. of probesNo. of casesCandidates
Amp7q22.199852752100767476425MUC17
7p22.3-p21.352493574289101283FSCN1
8q24.31452108371457820382411FOXH1
8q22.19506152997342088454CDH17
8p11.23393780513946183438ADAM5P, ADAM3A
8p11.21428169424284918634THAP1, RNF170
13q12.22709535227439560114CDX2
13q14.2-q14.346790942518991571174ALG11
13q31.3910754769117696044GPC5
16p11.22989092931412127784MAPK3
19p13.21114155711548932293CNN1
20q11.21-q11.23295015353501420115510
20q13.336000866062320720856PTK6, TNFRSF6B
20p11.231869242920864752423
HD5q14.38772262190788169483MEF2C
8p23.3-p21.2211611271996114673DLC1, PCM1
15q11.21883566020010618125
16p13.26492886686097293A2BP1
17p13.1-p121108988015073870693MAP2K4
18q21.2467647964710776493SMAD4
18q21.2480230004842362783DCC
20p12.1147723721493955244MACROD2
Genomic changes associated with lymph node metastasis, tumor stage and distant metastasis

We compared the frequencies of genomic aberrations in the rectal cancer patients subdivided accoding to cases with or without lymph node metastasis, with tumor stages II or III-IV, and with or without distant metastasis using MD-SeeGH software. The results showed that gains of 7p, 13q and losses of 4p, 4q and 8p were more frequent in the rectal cancers with lymph node metastasis (Fig. 2A). The frequencies of 7p, 13q gain and 4q loss were higher in stage III-IV cases when compared with the frequencies in stage II cases (Fig. 2B). The largest differences were detected in copy number changes of 4q and 17q, with more frequent 4q loss and 17q gain in the cases with distant metastasis (Fig. 2C). We analyzed these candidate genomic regions with clinical factors by Chi-square test, and found that losses of 4p16.1-p15.31, 8p21.1-p12 and gains of 7p12.3-p12.1 and 13q33.1-q34 were associated with positive lymph node metastasis and advanced clinical stage (stages III and IV). Moreover, loss of 4q34.3-q35.1 was linked only with advanced stage (stages III and IV). We also found that the patients with distant metastasis had more frequent 17q24.2–25.3 gain (Table III).

Table III

Genomic aberrations linked with clinicopathological characteristics of the rectal cases.

Table III

Genomic aberrations linked with clinicopathological characteristics of the rectal cases.

pN statusDistant metastasisStage



CytobandChangePositiveNegativeP-valuePositiveNegativeP-valueStage IIStages III and IVP-value
4p16.1-p15.31Loss920.036470.4301100.013
No loss17209281918
4q34.3-q35.1Loss1140.072690.1753120.040
No loss15187261716
7p12.3-p12.1Gain1210.001490.7261120.004
No gain14219261916
8p21.1-p12Loss1650.0075160.6535160.027
No loss10178191512
13q33.1-q34Gain1330.0083130.3583130.023
No gain131910221715
17q24.2-q25.3Gain440.796620.001260.295
No gain22187331822
Candidate target genes of gains and losses in rectal carcinoma

We performed an integrated analysis of the array-CGH dataset and the gene expression profiling dataset of CRC cells of NCI-60 to identify the candidate target genes of genomic gains and losses. The expression level of CA2, PDP1, ANGPT1, LOC346887, MAL2, NOV, TRIB1 and ZNF572 was higher in cell lines with an 8q gain than without. We also analyzed candidate target genes of gains in 13q, 17q, 20p and 20q and of loss in 18q (Table IV) and found that PDP1 (8q), TRIB1 (8q), C13orf27 (13q), FOXA2 (20p), PMEPA1 (20q) and PHACTR3 (20q) were overexpressed not only in CRC but also in other types of cancers (Table V). Three genes in 18q (FHOD, SMAD4 and BCL2) presented underexpression in several types of cancers including CRC (Table V).

Table IV

Candidate target genes of gains and losses in rectal cancer.

Table IV

Candidate target genes of gains and losses in rectal cancer.

ChangeCytobandGenes (>5-fold change)
Gain8qCA2, PDP1, ANGPT1, LOC346887, MAL2, NOV, TRIB1, ZNF572
13qOXGR1, PCDH9, EFNB2, C13orf27, LMO7, ARL11
17qTTYH2, RAB37, MXRA7, SLC26A11, MGAT5B
20pFOXA2, C20orf56, SLC24A3, CHGB, C20orf194, NRSN2
20qPMEPA1, PCK1, SULF2, LOC100240735, WFDC2, PHACTR3
Loss8pNot found
11qNot found
18qFHOD3, PSTPIP2, SMAD4, MBD2, BCL2, ST8SIA5

Table V

Candidate targets in the Oncomine database.

Table V

Candidate targets in the Oncomine database.

Colorectal cancer

CytobandGeneUpDownChange in other cancers
8q gainPDP180Cervical cancer, gastic cancer, head and neck cancer, kidney cancer, leukemia, lymphoma, melanoma, pancreatic cancer
TRIB110Brain and CNS cancer, breast cancer, esophageal cancer, head and neck cancer, leukemia, lymphoma, melanoma, prostate cancer
ZNF57220No
13q gainOXGR141No
C13orf2750Cervical cancer
20p gainFOXA261Esophageal cancer
20q gainPMEPA190Bladder cancer, breast cancer, esophageal cancer, gastric cancer, head and neck cancer, lung cancer, pancreatic cancer
PHACTR320Brain and CNS cancer, pancreatic cancer
18q lossFHOD301Bladder cancer, brain and CNS cancer, breast cancer, kidney cancer, prostate cancer
SMAD402Lymphoma
BCL2014Bladder cancer, brain and CNS cancer, breast cancer, head and neck cancer, leukemia, lymphoma, ovarian cancer, prostate cancer, sarcoma
Pathways enriched for copy number alterations

Pathway enrichment analysis using KEGG database was applied to the CGH data, and we found four pathways enriched in genes with gain and three pathways enriched in genes with loss. The genomic gains in rectal carcinoma changed the pathways of nitrogen metabolism, oxidative phosphorylation, cell cycle and maturity onset diabetes of young. However, cytokine-cytokine receptor interaction, MAPK signaling pathway, and dentatorubropallidoluysian atrophy (DRPLA) pathways were changed by the genomic losses (Table VI).

Table VI

Pathways enriched in the array-CGH data.

Table VI

Pathways enriched in the array-CGH data.

AberrationPathwayDescriptionNo. of genesGene symbolP-value
Gainhsa00910Nitrogen metabolism22CA6, CTH, CA14, GLUL, GLS, CPS1, AMT, ASNS, CA8, CA13, CA1, CA3, CA2, CA9, GLUD1, GLS2, HAL, CA12, CA7, CA5A, CA4, CA110.003
Gainhsa00190Oxidative phosphorylation109SDHB, NDUFS5, ATP6V0B, UQCRH, ATP5F1, NDUFS2, SDHC, ATP6V1G3, ATP6V1C2, COX7A2L0.017
Gainhsa04110Cell cycle108MAD2L2, SFN, HDAC1, CDC20, CDKN2C, ORC1, GADD45A, CDC7, CDC14A, TGFB20.025
Gainhsa04950Maturity onset diabetes of the young24PKLR, NR5A2, NEUROD1, SLC2A2, HES1, NKX6-1, GCK, BHLHA15, PAX4, MNX1, HNF4G, MAFA, NEUROG3, HHEX, PAX6, IAPP, HNF1A, PDX1, ONECUT1, HNF1B, FOXA3, NKX2-2, FOXA2, HNF4A0.044
Losshsa04060Cytokine-cytokine receptor interaction240TNFRSF18, TNFRSF4, TNFRSF14, TNFRSF25, TNFRSF9, TNFRSF8, TNFRSF1B, IL22RA1, IL28RA, CSF3R0.001
Losshsa04010MAPK signaling pathway256CASP9, PLA2G2E, PLA2G2A, PLA2G5, PLA2G2D, PLA2G2F, CDC42, STMN1, RPS6KA1, MAP3K60.011
Losshsa05050 Dentatorubropallidoluysian atrophy (DRPLA)14RERE, CASP8, MAGI1, CASP3, MAGI2, WWP1, CASP7, CASP1, GAPDH, ATN1, WWP2, BAIAP2, INSR, ITCH0.038

Discussion

The biological properties of cancers are different in patients presenting with different clinical parameters such as invasive depth, lymph node metastasis, distant metastasis, differentiation and clinical stage. Thus, the optimal treatment should be based on an individual cancer. Biomarkers can improve the accuracy of determining the clinical parameters that are predictors of prognosis and indicators of a response to treatment.

By applying array-CGH to rectal carcinoma samples of Chinese parients, we screened the genomic aberrations associated with clinical parameters using frequency comparison. The results showed that losses of 4p16.1-p15.31, 8p21.1-p12 and gains of 7p12.3-p12.1 and 13q33.1-q34 were associated with positive lymph node metastasis and advanced clinical stage (stages III and IV). Loss of 4q34.3-q35.1 was a marker of advanced stage (stages III and IV). We also found that the patients with distant metastasis had a more frequent gain in 17q24.2–25.3. Chromosome 4 was found to be the most frequent loss region in cancers including cervical, esophageal, lung, head and neck, gastric and CRC (1116). The incidence of 4q loss was much higher in pulmonary metastatic tissues when compared with primary cancer tissues and LOH of the D4S1534 locus (4q) in primary tissues was significantly linked with liver metastasis (17,18). Our results also suggested that loss of 4q in primary rectal tissues was a candidate predictor of lymph node metastasis. This indicates that target genes of the loss of 4q play important roles in lymphatic invasion and tumor progression, to which further investigation should be addressed.

The correlation of 17q with clinicopathological factors of CRC was not explicit. Diep et al(7) found that gain of 17q was correlated with the transition from a primary tumor to liver metastasis, while Knosel et al(16) reported that more deletion at 17q were observed in lung metastasis than primary tumors (19). Our results showed that the gain of 17q was more frequent in rectal cancer patients with distant metastasis when compared with patients without metastasis. Additional independent validation assays should be performed to reveal the correlation of 17q and metastasis.

Our results revealed that the alteration in expression of PDP1 (8q), TRIB1 (8q), C13orf27 (13q), FOXA2 (20p), PMEPA1 (20q), PHACTR3 (20q), FHOD (18q), SMAD4 (18q) and BCL2 (18q) occurred in CRC and other types of cancer, with a consistent copy number increase or decrease. To date, there is no report concerning the function of PDP1, C13orf27 and FHOD in cancer. Our results indicate the need to study these genes in rectal carcinogenesis. TRIB1 is a mammalian homolog of tribbles, an evolutionarily conserved Drosophila protein family that regulates protein degradation. In myeloid leukemogenesis, TRIB1 was found to be overexpressed and was a key mediator between the RTK-MAPK pathway and the C/EBP transcription factor (20,21). FOXA2 was found to function as a suppressor of tumor metastasis by inhibition of epithelial-to-mesenchymal transition (EMT). Loss of FOXA2 expression due to epigenetic silencing was frequent in lung cancer (22,23). PMEPA1 is a TGF-β inducible gene and encodes an NEDD4 E3 ubiquitin ligase binding protein. PMEPA1 was found to be overexpressed in prostate, breast, renal cell, stomach and rectal carcinomas (2426). PHACTR3 was identified as a PP1-binding protein and was selectively expressed in the brain. PHACTR3 was found to be overexpressed in 20% of non-small cell lung cancer (NSCLC), and was associated with reduced survival time of patients. In advanced neoplasia the methylation level of PHACTR3 was 70-fold higher than that in normal colon mucosa, and the sensitivity and specificity in stool assay were 55 and 95%, respectively (27). Loss of SMAD4 expression was reported as a predictor of liver metastasis in CRC, and patients with reduced SMAD4 expression presented with a poor prognosis (2830). Transgenic expression of SMAD4 was found to significantly reduce the oncogenic potential of SW620 and MC38 cell lines (31). Overall, these genes may be the target genes of genomic gains and losses in rectal cancer. Further research should be addressed to elucidate the roles of the candidate genes in rectal carcinogenesis.

In summary, the genomic aberrations identified in the present study can be suggested as candidate biomarkers with which to predict the clinical outcome of patients with rectal carcinoma and may be expected to serve to individualize the treatment of rectal cancer. Our study identified several candidate target genes of the most common gains and losses in rectal cancer, and our findings provide information to explore the role of these genes in the development and progression of rectal cancer.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (30950013) and the Special Public Health Fund of China (200902002-5).

References

1 

Ministry of Health, PCR. Chinese Health Statistical Digest. 2010, Available from: http://www.moh.gov.cn/publicfiles//business/htmlfiles/zwgkzt/ptjty/digest2010/index.html.

2 

Alazzouzi H, Alhopuro P, Salovaara R, Sammalkorpi H, Jarvinen H, Mecklin JP, Hemminki A, Schwartz S Jr, Aaltonen LA and Arango D: SMAD4 as a prognostic marker in colorectal cancer. Clin Cancer Res. 11:2606–2611. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Basseres DS, D’Alo F, Yeap BY, Lowenberg EC, Gonzalez DA, Yasuda H, Dayaram T, Kocher ON, Godleski JJ, Richards WG, Meyerson M, Kobayashi S, Tenen DG, Halmos B and Costa DB: Frequent downregulation of the transcription factor Foxa2 in lung cancer through epigenetic silencing. Lung Cancer. 77:31–37. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Bockmuhl U, Schmidt S, Petersen S and Petersen I: Deletion of chromosome 10q - a marker for metastasis of head-neck carcinomas? Laryngorhinootologie. 79:81–85. 2000.(In German).

5 

Bosch LJ, Oort FA, Neerincx M, Khalid-de Bakker CA, Terhaar sive Droste JS, Melotte V, Jonkers DM, Masclee AA, Mongera S, Grooteclaes M, Louwagie J, van Criekinge W, Coupe VM, Mulder CJ, van Engeland M, Carvalho B and Meijer GA: DNA methylation of phosphatase and actin regulator 3 detects colorectal cancer in stool and complements FIT. Cancer Prev Res. 5:464–472. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Dellas A, Torhorst J, Jiang F, Proffitt J, Schultheiss E, Holzgreve W, Sauter G, Mihatsch MJ and Moch H: Prognostic value of genomic alterations in invasive cervical squamous cell carcinoma of clinical stage IB detected by comparative genomic hybridization. Cancer Res. 59:3475–3479. 1999.PubMed/NCBI

7 

Diep CB, Kleivi K, Ribeiro FR, Teixeira MR, Lindgjaerde OC and Lothe RA: The order of genetic events associated with colorectal cancer progression inferred from meta-analysis of copy number changes. Genes Chromosomes Cancer. 45:31–41. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Fearon ER and Vogelstein B: A genetic model for colorectal tumorigenesis. Cell. 61:759–767. 1990. View Article : Google Scholar : PubMed/NCBI

9 

Giannini G, Ambrosini MI, Di Marcotullio L, Cerignoli F, Zani M, MacKay AR, Screpanti I, Frati L and Gulino A: EGF- and cell-cycle-regulated STAG1/PMEPA1/ERG1.2 belongs to a conserved gene family and is overexpressed and amplified in breast and ovarian cancer. Mol Carcinog. 38:188–200. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Hu N, Roth MJ, Emmert-Buck MR, Tang ZZ, Polymeropolous M, Wang QH, Goldstein AM, Han XY, Dawsey SM, Ding T, Giffen C and Taylor PR: Allelic loss in esophageal squamous cell carcinoma patients with and without family history of upper gastrointestinal tract cancer. Clin Cancer Res. 5:3476–3482. 1999.PubMed/NCBI

11 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar

12 

Jiang JK, Chen YJ, Lin CH, Yu IT and Lin JK: Genetic changes and clonality relationship between primary colorectal cancers and their pulmonary metastases - an analysis by comparative genomic hybridization. Genes Chromosomes Cancer. 43:25–36. 2005. View Article : Google Scholar

13 

Jiang LX, Xu J, Wang ZW, Li DP, Peng ZH, Gao JJ, He L and Zheng HT: Tumor suppress genes screening analysis on 4q in sporadic colorectal carcinoma. World J Gastroenterol. 14:5606–5611. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Kawakami M, Yamaguchi T, Takahashi K, Matsumoto H, Yasutome M, Horiguchi S, Hayashi Y, Funata N and Mori T: Assessment of SMAD4, p53, and Ki-67 alterations as a predictor of liver metastasis in human colorectal cancer. Surg Today. 40:245–250. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Kimura Y, Noguchi T, Kawahara K, Kashima K, Daa T and Yokoyama S: 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. 17:1328–1337. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Knosel T, Schluns K, Dietel M and Petersen I: Chromosomal alterations in lung metastases of colorectal carcinomas: associations with tissue specific tumor dissemination. Clin Exp Metastasis. 22:533–538. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Lagerstedt KK, Staaf J, Jonsson G, Hansson E, Lonnroth C, Kressner U, Lindstrom L, Nordgren S, Borg A and Lundholm K: Tumor genome wide DNA alterations assessed by array CGH in patients with poor and excellent survival following operation for colorectal cancer. Cancer Inform. 3:341–355. 2007.PubMed/NCBI

18 

Lei T, Chen WQ, Zhang SW, Lei TH, Ying Q, He ZY and Wang XH: Prevalence trend of colorectal cancer in 10 cities and counties in China from 1988 to 2002. Zhonghua Zhong Liu Za Zhi. 31:428–433. 2009.(In Chinese).

19 

Lengauer C, Kinzler KW and Vogelstein B: Genetic instability in colorectal cancers. Nature. 386:623–627. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Lengauer C, Kinzler KW and Vogelstein B: Genetic instabilities in human cancers. Nature. 396:643–649. 1998. View Article : Google Scholar : PubMed/NCBI

21 

Li HL, Gao YT, Zheng Y, Zhang W, Gao LF, Xu B and Xiang YB: Incidence trends of colorectal cancer in urban Shanghai, 1973–2005. Zhonghua Yu Fang Yi Xue Za Zhi. 43:875–879. 2009.(In Chinese).

22 

Losi L, Bouzourene H and Benhattar J: Loss of Smad4 expression predicts liver metastasis in human colorectal cancer. Oncol Rep. 17:1095–1099. 2007.PubMed/NCBI

23 

Nakao M, Kawauchi S, Furuya T, Uchiyama T, Adachi J, Okada T, Ikemoto K, Oga A and Sasaki K: Identification of DNA copy number aberrations associated with metastases of colorectal cancer using array CGH profiles. Cancer Genet Cytogenet. 188:70–76. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Nakao M, Kawauchi S, Uchiyama T, Adachi J, Ito H, Chochi Y, Furuya T, Oga A and Sasaki K: DNA copy number aberrations associated with the clinicopathological features of colorectal cancers: Identification of genomic biomarkers by array-based comparative genomic hybridization. Oncol Rep. 25:1603–1611. 2011.

25 

Petersen S, Aninat-Meyer M, Schluns K, Gellert K, Dietel M and Petersen I: Chromosomal alterations in the clonal evolution to the metastatic stage of squamous cell carcinomas of the lung. Br J Cancer. 82:65–73. 2000.PubMed/NCBI

26 

Rae FK, Hooper JD, Nicol DL and Clements JA: Characterization of a novel gene, STAG1/PMEPA1, upregulated in renal cell carcinoma and other solid tumors. Mol Carcinog. 32:44–53. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Rothlisberger B, Heizmann M, Bargetzi MJ and Huber AR: TRIB1 overexpression in acute myeloid leukemia. Cancer Genet Cytogenet. 176:58–60. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Tang Y, Shu G, Yuan X, Jing N and Song J: FOXA2 functions as a suppressor of tumor metastasis by inhibition of epithelial-to-mesenchymal transition in human lung cancers. Cell Res. 21:316–326. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Xu LL, Shanmugam N, Segawa T, Sesterhenn IA, McLeod DG, Moul JW and Srivastava S: A novel androgen-regulated gene, PMEPA1, located on chromosome 20q13 exhibits high level expression in prostate. Genomics. 66:257–263. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Yokoyama T, Kanno Y, Yamazaki Y, Takahara T, Miyata S and Nakamura T: Trib1 links the MEK1/ERK pathway in myeloid leukemogenesis. Blood. 116:2768–2775. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Zhang B, Halder SK, Kashikar ND, Cho YJ, Datta A, Gorden DL and Datta PK: Antimetastatic role of Smad4 signaling in colorectal cancer. Gastroenterology. 138:969–980. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May 2013
Volume 29 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liang J, Shi Z, Zhang T, Hao J, Wang Z, Wang X, Yang H, Wang M, Zhou Z, Zhang Y, Zhang Y, et al: Analysis of genomic aberrations associated with the clinicopathological parameters of rectal cancer by array‑based comparative genomic hybridization. Oncol Rep 29: 1827-1834, 2013
APA
Liang, J., Shi, Z., Zhang, T., Hao, J., Wang, Z., Wang, X. ... Zhang, Y. (2013). Analysis of genomic aberrations associated with the clinicopathological parameters of rectal cancer by array‑based comparative genomic hybridization. Oncology Reports, 29, 1827-1834. https://doi.org/10.3892/or.2013.2296
MLA
Liang, J., Shi, Z., Zhang, T., Hao, J., Wang, Z., Wang, X., Yang, H., Wang, M., Zhou, Z., Zhang, Y."Analysis of genomic aberrations associated with the clinicopathological parameters of rectal cancer by array‑based comparative genomic hybridization". Oncology Reports 29.5 (2013): 1827-1834.
Chicago
Liang, J., Shi, Z., Zhang, T., Hao, J., Wang, Z., Wang, X., Yang, H., Wang, M., Zhou, Z., Zhang, Y."Analysis of genomic aberrations associated with the clinicopathological parameters of rectal cancer by array‑based comparative genomic hybridization". Oncology Reports 29, no. 5 (2013): 1827-1834. https://doi.org/10.3892/or.2013.2296