Open Access

Integrin α‑5 as a potential biomarker of head and neck squamous cell carcinoma

  • Authors:
    • Bo Zou
    • Dong Wang
    • Kai Xu
    • Dao‑Ying Yuan
    • Zhen Meng
    • Bin Zhang
  • View Affiliations

  • Published online on: August 22, 2019     https://doi.org/10.3892/ol.2019.10773
  • Pages: 4048-4055
  • Copyright: © Zou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Head and neck squamous cell carcinoma (HNSCC) is one of the most common malignant tumors that endanger human health. In recent years, the incidence of HNSCC has been increasing, without any significant improvement in the prognosis. Therefore, increased knowledge on the molecular mechanism underlying HNSCC development will allow the development of new strategies for therapy. The present study attempted to identify key genes involved in HNSCC development. Expression profiles of HNSCC and normal samples were downloaded from The Cancer Genome Atlas database. Differentially expressed genes (DEGs) between the HNSCC and normal samples were identified and subjected to Gene Oncology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analysis. A protein‑protein interaction (PPI) network was constructed, and Cytoscape CentiScape and Gene Expression Profiling Interactive Analysis were used to identify key DEGs. Finally, expression profiles of HNSCCs, including 500 HNSCCs and 44 normal samples, were included in the analysis. A total of 1,181 DEGs were screened, among which 354 genes were upregulated and 827 genes were downregulated in HNSCC compared with normal tissues. The GO enrichment analysis showed that the DEGs were mainly involved in chloride transmembrane transporter, metalloendopeptidase and substrate‑specific channel activities. The KEGG pathway analysis revealed that the DEGs were mainly associated with ‘protein digestion and absorption’, as well as ‘extracellular matrix‑receptor interaction’. Integrin α‑5 (ITGA5) was identified as a hub gene, based on the PPI network complex, and was confirmed to be significantly associated with the overall survival rate. Moreover, ITGA5 was overexpressed specifically in HNSCC. The genes found, notably ITGA5, are potential diagnostic biomarkers and therapeutic targets in HNSCC.

Introduction

Head and neck squamous cell carcinoma (HNSCC) was reported as the sixth most common type of malignancy in humans, constituting ~4% of all new cases in the United States in 2015 (1,2). According to a recent report (2018), ~600,000 patients were affected worldwide yearly and the incidence rate has significantly increased (3). Improvements in clinical therapy have not led to corresponding improvements in the prognosis of patients with HNSCC. The 5-year survival rate of patients with HNSCC still remains between 40 and 50%. Revealing the underlying mechanism of HNSCC development could provide potential biomarkers or therapeutic targets in HNSCC (4).

Several studies have focused on the mechanism of HNSCC, and the new generation of sequencing technology provides a rich resource for the study of significant genetic changes during tumorigenesis and for the screening of potential diagnostic and prognostic markers of cancer. For instance, it was reported that actin-like protein 8 (ACTL8) was increasingly expressed in HNSCC and regarded as an independent prognostic factor (5). The expression of neutrophil gelatinase-associated lipocalin was lower in HNSCC than in normal tissues, and was correlated with the tumorigenesis of HNSCC (6). Calpain 6 expression was significantly decreased in HNSCC and positively associated with the survival rate of patients with the disease, thereby indicating the role of calpain 6 as a tumor suppressor in HNSCC (7). However, due to the limited sample sizes, previous studies may provide false predictions.

In the present study, integrated analysis was performed to identify the key genes involved in the development of HNSCC. Firstly, the differentially expressed genes (DEGs) between HNSCC and normal tissues were screened, followed by Gene Ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis and protein-protein interaction (PPI) network analysis. Finally, the key candidate DEGs were identified according to Centiscape and log-rank survival analysis, and were verified using the Gene Expression Ontology (GEO) datasets. These key DEGs were identified as potential biomarkers for early diagnosis and as therapeutic targets for HNSCC.

Materials and methods

Gene expression profile data and identification of DEGs

The level-3 RNA sequence (RNA Seq) data (fragments per kilobase of transcript per million mapped reads upper quartile data) of HNSCC and corresponding normal tissue samples were downloaded from the TCGA database (dataset no. :544; http://www.cancer.gov/tcga) (8) using the Genomic Data Commons Application Programming Interface (9). A total of 544 samples from 500 patients with HNSCC and 44 normal controls were collected in December 2018. The normal controls included normal tissues from the oral cavity, oral tongue, larynx, floor of the mouth and base of the tongue. The raw data was downloaded and the log2 fold-change (log2FC) was calculated using the Limma R package (version 3.2.5; http://www.r-project.org/) to screen DEGs between HNSCC and normal tissues. The following cut-off criteria were applied: log2FC>2 and P<0.05. The adjustment of P<0.5 was set as the threshold to adjust the P-value for multiple comparisons.

For verification purposes, the microarray expression dataset GSE6631 was downloaded (as minimum information about a microarray experiment notation in mark-up language formatted family files) from the GEO database (10). The GSE6631 was based on the GPL8300 Platforms (Affymetrix Human Genome U95 version 2 array) and included 44 HNCC samples and paired normal samples (submission date, 2007; last updated, 2018) (11). The sample information and expression profile data were extracted by R package (version:3.2.5) from GES6631. Statistical analyses were performed with GraphPad Prism version 8.0 software (GraphPad Software, Inc.). Single comparisons between two groups were performed using Paired Student's t-test.

GO and pathway enrichment analyses of DEGs

GO analysis and KEGG pathway enrichment of DEGs were performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID) (version, 6.7; http://david-d.ncifcrf.gov/) to screen for possible biological processes, cellular components, molecular functions and signaling pathways of the involved DEGs (12). The resulting data were imported into Cytoscape ClueGo software (version: 3.6.0) for visual analysis (13). P<0.05 was considered as statistically significant. The following parameter settings were applied: Identifier, ‘official gene symbol.’; list type, ‘gene list.’; species, ‘homo sapiens.’; count threshold, 2; and ease threshold, 0.05.

PPI network construction and candidate gene identification

The Search Tool for the Retrieval of Interacting Genes/Proteins (STRING; version, 11.0; http://string-db.org) was used to construct the PPI network (14). The minimum required interaction score was set to a medium confidence of 0.4, and the organism was set to ‘Homo sapiens’. The Cytoscape software was then used to visualize the network. Cytoscape CentiScape (version, 3.6.0; http://apps.cytoscape.org/apps/centiscape) (15) was used to screen candidate key proteins in the network, according to the degree of centrality. The genes with a node degree of ≥15 were considered as the candidate key genes.

Association between candidate key genes and clinicopathological parameters of HNSCC

By using the Gene Expression Profiling Interactive Analysis (GEPIA) database (http://gepia.cancer-pku.cn/), the effect of candidate key genes on overall survival (OS) rate was evaluated using log-rank test and the Mantel-Cox test. P<0.05 for log-rank test indicated statistical significance. Genes significantly associated with HNSCC OS rate were considered as key genes. The following parameter settings were applied: Group cut-off, ‘median.’; hazards ratio, ‘yes.’; 95% confidence interval, ‘yes.’; and axis units, ‘months.’.

Comparison of key genes in HNSCC and other types of cancer

In order to evaluate the specificity of the key genes screened in the previous step to HNSCC, the expression of these genes was examined in other tumor datasets from the GEPIA database, including adrenocortical carcinoma, bladder urothelial carcinoma (BLCA), breast invasive carcinoma, endocervical adenocarcinoma (CESC), colon adenocarcinoma (COAD), esophageal carcinoma (ESCA), liver hepatocellular carcinoma, lung squamous cell carcinoma and lung adenocarcinoma (LUAD). These datasets came from the TCGA and the GTEx projects (16). The unmatched normal and tumor tissues were compared. The raw data were filtered based on the cut-offs log2FC>2 and P<0.05.

Results

Identification of DEGs in HNSCC

RNAseq data of HNSCC and corresponding normal tissue samples were downloaded from the TCGA database. The data was screened by the Limma package, using P<0.05 and log2FC>2 as the cut-off criteria, which identified 1,181 DEGs (Fig. 1), including 354 upregulated and 827 downregulated genes.

GO analysis and signaling pathway enrichment of DEGs in HNSCC

The GO analysis of the 1,181 DEGs was performed using the DAVID database, with the criterion set at P<0.05. The DEGs were divided into three groups, namely, biological process, cellular component and molecular function groups. As shown in Fig. 2A, the main DEG-associated biological functions were ‘cell adhesion’, ‘extracellular matrix organization’, ‘skeletal system development’ and ‘ion transmembrane transport’. As demonstrated in Fig. 2B, the cellular component analysis revealed that the selected DEGs were mainly located at the ‘extracellular exosome’, ‘extracellular region’ and ‘extracellular space’. The molecular function of the DEGs was mainly associated with ‘actin binding’, ‘heparin binding’ and ‘cytokine activity’ (Fig. 2C). As demonstrated in Fig. 3, the KEGG pathways enriched by the DEGs were mainly associated with ‘protein digestion and absorption’, ‘extracellular matrix-receptor interaction’, ‘drug metabolism’ and the ‘PPAR signaling pathway’.

Key candidate DEG identification with PPI network analysis

The PPI network of the DEG expression products was constructed using Cytoscape software and the STRING database (http://string-db.org). A total of 1,035 DEGs were incorporated into the PPI network complex. The Cytoscape CentiScape was used to screen candidate key genes in the network, with degree of centrality ≥15 set as the inclusion criterion, which identified 50 genes that were included in the following analysis (Fig. 4).

Log-rank survival analysis by the GEPIA database, found 8 out of the 50 candidate key genes, including integrin α-5 (ITGA5) and serpin family E member 1 (SERPINE1), to be significantly associated with HNSCC OS rate (P<0.05; Table I). If the threshold was set to P<0.01, only ITGA5 and SERPINE1 were found to be associated with the OS rate of HNSCC (Fig. 5). The Cox proportional hazard ratios of ITGA5 and SERPINE1 were both 1.5.

Table I.

Log regression analysis of differently expressed genes with node degree of centrality ≥15.

Table I.

Log regression analysis of differently expressed genes with node degree of centrality ≥15.

Gene symbolDegree of centralityLog-rank, P-value
COL1A1450.30
COL1A2370.84
FN1340.11
COL2A1340.26
COL3A1330.56
MMP9320.93
COL4A2300.61
COL4A1300.62
COL5A2290.29
COL4A5280.85
COL11A1260.09
COL7A1260.10
COL5A1260.28
SPARC260.30
COL6A3260.86
SERPINH1250.02
COL6A1250.22
COL4A6250.36
TIMP1240.04
SPP1240.04
CSF2230.02
LUM230.63
COL10A1230.98
GNGT1220.15
COL27A1220.46
ITGA521<0.01
MMP1210.04
CXCL10210.19
CENPA210.30
COL12A1210.53
P4HA3200.06
MMP13200.09
MMP3200.23
POSTN200.30
PLK1200.41
COL13A1200.44
COL22A1200.62
FOXM1190.55
ACAN180.50
AURKB180.63
KIF2C170.29
CDC45170.90
PTHLH170.92
SERPINE116<0.01
ITGA11160.50
TPX2160.61
OASL160.67
NID1150.04
CXCL9150.18
UBE2C150.58
ITGA5 is highly expressed in HNSCC specifically

To evaluate the specificity of the key genes previously screened in HNSCC, their expression levels were determined in other tumor datasets from the GEPIA database. The expression of ITGA5 was only significantly increased in HNSCC and decreased in BLCA, CESC, COAD and LUSC (Fig. 6A). However, SERPINE1 expression was significantly increased in ESCA, as well as in HNSCC, with no other significant changes in other tumors (Fig. 6B).

For verification, ITGA5 expression was evaluated in HNSCC based on the microarray express dataset GSE6631. Compared with ITGA5 expression in normal tissues, the expression in HNSCC tissues was upregulated (P=0.007; Fig. 7).

Discussion

HNSCC is one of the most common types of malignancies in humans; it is characterized by rapid progression, a high migration capacity and high mortality rate. However, there are almost no biomarkers or targets for the diagnosis and treatment of HNSCC (1). Thus, the identification of genes that are differentially expressed between tumor and normal tissues will contribute to future studies on the pathogenesis of HNSCC and will provide potential diagnostic biomarkers and therapeutic targets.

In the present study, TCGA datasets of HNSCC were integrated and analyzed using bioinformatics. Finally, a total of 1,181 DEGs were identified including 354 upregulated genes and 827 downregulated genes in the initial step. Subsequently, the 1,181 DEGs were analyzed based on GO terms and classified based on KEGG signaling pathways. The GO analysis indicated that the DEGs were mainly involved in ‘cell adhesion’, ‘extracellular matrix organization’, ‘skeletal system development’, ‘ion transmembrane transport’, ‘actin binding’, ‘heparin binding’ and ‘cytokine activity’. The KEGG pathway analysis revealed that the DEGs were mainly associated with ‘protein digestion and absorption’, ‘extracellular matrix-receptor interaction’, ‘drug metabolism’ and the ‘PPAR signaling pathway’. In addition, the PPI network complex was constructed and eight key genes, including ITGA5, SERPINE1, serpin family H member 1, colony-stimulating factor 2, tissue inhibitor of metalloproteinases 1, nidogen 1, secreted phosphoprotein 1 and matrix metallopeptidase 1, were screened based on centrality and log-rank survival analysis by GEPIA. The GEPIA database contains genotype-tissue expression data, which increases the sample size and accuracy of the analysis, and was therefore was used to identify key DEGs. As a result, ITGA5 and SERPINE1 were identified as key genes.

ITGA5 is an important member of the integrin family, and its coding gene is located at the human chromosome 12q11-q13. Integrin is an extracellular matrix receptor that acts as an adhesive receptor for extracellular matrix proteins, including fibrin, laminin and collagen (17). ITGA5 forms the link between the extracellular matrix and intracellular signal transduction, and also participates in a variety of important physiological processes; it is also associated with tumor occurrence, development, invasion and metastasis (18). A study on hepatocellular carcinoma (HCC) demonstrated a negative correlation between the expression of ITGA5 and miR-128, and the downregulation of ITGA5 leading to the inhibition of HCC cell metastasis and stem cell-like properties (19). However, the expression of ITGA5 played diverse roles in breast cancer cells (20). Expression in highly invasive breast cancer cells was almost absent in comparison with that in less invasive cells, thereby indicating the negative association between ITGA5 and breast cancer metastasis (21,22). In ovarian cancer cells, downregulation of ITGA5 induced by forced expression of miR-17 significantly limited the adhesion, invasion and tumorigenesis ability of cancer cells (23). ITGA5 was reported to be highly expressed in glioblastoma compared with normal brain glial cells, and its downregulation inhibited proliferation, invasion and migration (24). ITGA5 expression was also upregulated in oral squamous cell carcinoma (OSCC), a common type of HNSCC (25). Knockdown of ITGA5 inhibited the proliferation, migration and invasion of OSCC cells, thereby indicating that ITGA5 could promote OSCC progression.

SERPINE1, also known as plasminogen activator inhibitor type I, is a primary inhibitor of plasminogen activators and a marker of poor prognosis in cancer. In colorectal cancer, SERPINE1 expression was upregulated and was significantly associated with grading and microsatellite instability (26). In esophageal cancer, SERPINE1 expression was upregulated and significantly associated with age range, which was consistent with the analysis of the present study (27). SEPINE1 was highly expressed in oral carcinomas compared with in the matched tumor adjacent normal tissues (28), and its overexpression resulted in increased proliferation and tumor budding. Moreover, SEPINE1 was associated with poor progression-free and cancer-specific survival in patients with head and neck cancer (29,30).

In conclusion, analysis of GEPIA datasets demonstrated the association between high expression of ITGA5 and SERPINE1 and poor OS rate in patients with HNSCC, with identical hazard ratio for both genes. The investigation of these genes was conducted in other tumor datasets, in order to determine their specificity to HNSCC. The expression of ITGA5 was significantly increased in HNSCC only and decreased in BLCA, CESC, COAD and LUSC. On the other hand, the expression of SERPINE1 was significantly increased in ESCA, as well as in HNSCC. A limitation of the present study was that the gene expression in HNSCC was only analyzed by using the public database. In subsequent experiments, molecular biology and cell biology methods will be utilized to further verify these results. Overall, these findings suggest the potential of ITGA5 as a diagnostic or prognostic marker and as a therapeutic target in HNSCC.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Medicine and Health Science Technology Development Plan Project, Shandong Province (grant no. 2017WSA15041), the National Natural Science Foundation of China (grant nos. 81472530 and 81602374) and the Natural Science Foundation of Shandong Province (grant no. 2016ZRA15065).

Availability of data and materials

All data generated or analyzed during the present study are included in this published article.

Authors' contributions

BoZ performed data download, survival analysis and drafted the manuscript. DW and KX performed the survival analysis, GO analysis and pathway enrichment analysis. DY worked on PPI network construction. ZM analyzed the association between the candidate genes and prognosis, and also participated the design of the study. BiZ was the major contributor in designing the present study. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Cancer Genome Atlas Network, . Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 517:576–582. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Goossens N, Nakagawa S, Sun X and Hoshida Y: Cancer biomarker discovery and validation. Transl Cancer Res. 4:256–269. 2015.PubMed/NCBI

5 

Li B, Zhu J and Meng L: High expression of ACTL8 is poor prognosis and accelerates cell progression in head and neck squamous cell carcinoma. Mol Med Rep. 19:877–884. 2019.PubMed/NCBI

6 

Wang L, Chen C, Li F, Hua Q, Chen S, Xiao B, Dai M, Li M, Zheng A, Yu D, et al: Down-regulation of neutrophil gelatinase-associated lipocalin in head and neck squamous cell carcinoma correlated with tumorigenesis, not with metastasis. Int J Clin Exp Pathol. 8:8857–8868. 2015.PubMed/NCBI

7 

Xiang Y, Li F, Wang L, Zheng A, Zuo J, Li M, Wang Y, Xu Y, Chen C, Chen S, et al: Decreased calpain 6 expression is associated with tumorigenesis and poor prognosis in HNSCC. Oncol Lett. 13:2237–2243. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Cancer Genome Atlas Research Network, ; Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C and Stuart JM: The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet. 45:1113–1120. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Wilson S, Fitzsimons M, Ferguson M, Heath A, Jensen M, Miller J, Murphy MW, Porter J, Sahni H, Staudt L, et al: Developing cancer informatics applications and tools using the NCI genomic data commons API. Cancer Res. 77:e15–e18. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Wilhite SE and Barrett T: Strategies to explore functional genomics data sets in NCBI's GEO database. Methods Mol Biol. 802:41–53. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Kuriakose MA, Chen WT, He ZM, Sikora AG, Zhang P, Zhang ZY, Qiu WL, Hsu DF, McMunn-Coffran C, Brown SM, et al: Selection and validation of differentially expressed genes in head and neck cancer. Cell Mol Life Sci. 61:1372–1383. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Huang da W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, Fridman WH, Pagès F, Trajanoski Z and Galon J: ClueGO: A cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics. 25:1091–1093. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al: STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47:D607–D613. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Scardoni G, Tosadori G, Faizan M, Spoto F, Fabbri F and Laudanna C: Biological network analysis with CentiScaPe: Centralities and experimental dataset integration. F1000Res. 3:1392014. View Article : Google Scholar : PubMed/NCBI

16 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Meighan CM, Kelly VE, Krahe EC and Gaeta AJ: α integrin cytoplasmic tails can rescue the loss of Rho-family GTPase signaling in the C. elegans somatic gonad. Mech Dev. 136:111–122. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Morandi EM, Verstappen R, Zwierzina ME, Geley S, Pierer G and Ploner C: ITGAV and ITGA5 diversely regulate proliferation and adipogenic differentiation of human adipose derived stem cells. Sci Rep. 6:288892016. View Article : Google Scholar : PubMed/NCBI

19 

Zhao X, Wu Y and Lv Z: miR-128 modulates hepatocellular carcinoma by inhibition of ITGA2 and ITGA5 expression. Am J Transl Res. 7:1564–1573. 2015.PubMed/NCBI

20 

Fang Z, Yao W, Xiong Y, Zhang J, Liu L, Li J, Zhang C and Wan J: Functional elucidation and methylation-mediated downregulation of ITGA5 gene in breast cancer cell line MDA-MB-468. J Cell Biochem. 110:1130–1141. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Konstantinovsky S, Smith Y, Zilber S, Tuft Stavnes H, Becker AM, Nesland JM, Reich R and Davidson B: Breast carcinoma cells in primary tumors and effusions have different gene array profiles. J Oncol. 2010:9690842010. View Article : Google Scholar : PubMed/NCBI

22 

Xiao Y, Li Y, Tao H, Humphries B, Li A, Jiang Y, Yang C, Luo R and Wang Z: Integrin alpha5 down-regulation by miR-205 suppresses triple negative breast cancer stemness and metastasis by inhibiting the Src/Vav2/Rac1 pathway. Cancer Lett. 433:199–209. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Gong C, Yang Z, Wu F, Han L, Liu Y and Gong W: miR-17 inhibits ovarian cancer cell peritoneal metastasis by targeting ITGA5 and ITGB1. Oncol Rep. 36:2177–2183. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Feng L, Ma J, Ji H, Liu Y and Hu W: miR-330-5p suppresses glioblastoma cell proliferation and invasiveness through targeting ITGA5. Biosci Rep. 37:BSR201700192017. View Article : Google Scholar : PubMed/NCBI

25 

Fan QC, Tian H, Wang Y and Liu XB: Integrin-α5 promoted the progression of oral squamous cell carcinoma and modulated PI3K/AKT signaling pathway. Arch Oral Biol. 101:85–91. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Mazzoccoli G, Pazienza V, Panza A, Valvano MR, Benegiamo G, Vinciguerra M, Andriulli A and Piepoli A: ARNTL2 and SERPINE1: Potential biomarkers for tumor aggressiveness in colorectal cancer. J Cancer Res Clin Oncol. 138:501–511. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Klimczak-Bitner AA, Kordek R, Bitner J, Musial J and Szemraj J: Expression of MMP9, SERPINE1 and miR-134 as prognostic factors in esophageal cancer. Oncol Lett. 12:4133–4138. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Gao S, Nielsen BS, Krogdahl A, Sørensen JA, Tagesen J, Dabelsteen S, Dabelsteen E and Andreasen PA: Epigenetic alterations of the SERPINE1 gene in oral squamous cell carcinomas and normal oral mucosa. Genes Chromosomes Cancer. 49:526–538. 2010.PubMed/NCBI

29 

Pavón MA, Arroyo-Solera I, Téllez-Gabriel M, León X, Virós D, López M, Gallardo A, Céspedes MV, Casanova I, López-Pousa A, et al: Enhanced cell migration and apoptosis resistance may underlie the association between high SERPINE1 expression and poor outcome in head and neck carcinoma patients. Oncotarget. 6:29016–29033. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Arroyo-Solera I, Pavón MA, León X, López M, Gallardo A, Céspedes MV, Casanova I, Pallarès V, López-Pousa A, Mangues MA, et al: Effect of serpinE1 overexpression on the primary tumor and lymph node, and lung metastases in head and neck squamous cell carcinoma. Head Neck. 41:429–439. 2019.PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 18 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zou B, Wang D, Xu K, Yuan DY, Meng Z and Zhang B: Integrin α‑5 as a potential biomarker of head and neck squamous cell carcinoma. Oncol Lett 18: 4048-4055, 2019
APA
Zou, B., Wang, D., Xu, K., Yuan, D., Meng, Z., & Zhang, B. (2019). Integrin α‑5 as a potential biomarker of head and neck squamous cell carcinoma. Oncology Letters, 18, 4048-4055. https://doi.org/10.3892/ol.2019.10773
MLA
Zou, B., Wang, D., Xu, K., Yuan, D., Meng, Z., Zhang, B."Integrin α‑5 as a potential biomarker of head and neck squamous cell carcinoma". Oncology Letters 18.4 (2019): 4048-4055.
Chicago
Zou, B., Wang, D., Xu, K., Yuan, D., Meng, Z., Zhang, B."Integrin α‑5 as a potential biomarker of head and neck squamous cell carcinoma". Oncology Letters 18, no. 4 (2019): 4048-4055. https://doi.org/10.3892/ol.2019.10773