Skip to main content
Top
Published in: Cancer Cell International 1/2020

Open Access 01-12-2020 | Epigenetics | Primary research

Identification and validation of methylation-driven genes prognostic signature for recurrence of laryngeal squamous cell carcinoma by integrated bioinformatics analysis

Authors: Jie Cui, Liping Wang, Waisheng Zhong, Zhen Chen, Jie Chen, Hong Yang, Genglong Liu

Published in: Cancer Cell International | Issue 1/2020

Login to get access

Abstract

Background

Recurrence remains a major obstacle to long-term survival of laryngeal squamous cell carcinoma (LSCC). We conducted a genome-wide integrated analysis of methylation and the transcriptome to establish methylation-driven genes prognostic signature (MDGPS) to precisely predict recurrence probability and optimize therapeutic strategies for LSCC.

Methods

LSCC DNA methylation datasets and RNA sequencing (RNA-seq) dataset were acquired from the Cancer Genome Atlas (TCGA). MethylMix was applied to detect DNA methylation-driven genes (MDGs). By univariate and multivariate Cox regression analyses, five genes of DNA MDGs was developed a recurrence-free survival (RFS)-related MDGPS. The predictive accuracy and clinical value of the MDGPS were evaluated by receiver operating characteristic (ROC) and decision curve analysis (DCA), and compared with TNM stage system. Additionally, prognostic value of MDGPS was validated by external Gene Expression Omnibus (GEO) database. According to 5 MDGs, the candidate small molecules for LSCC were screen out by the CMap database. To strengthen the bioinformatics analysis results, 30 pairs of clinical samples were evaluated by digoxigenin-labeled chromogenic in situ hybridization (CISH).

Results

A total of 88 DNA MDGs were identified, and five RFS-related MDGs (LINC01354, CCDC8, PHYHD1, MAGEB2 and ZNF732) were chosen to construct a MDGPS. The MDGPS can effectively divide patients into high-risk and low-risk group, with the area under curve (AUC) of 0.738 (5-year RFS) and AUC of 0.74 (3-year RFS). Stratification analysis affirmed that the MDGPS was still a significant statistical prognostic model in subsets of patients with different clinical variables. Multivariate Cox regression analysis indicated the efficacy of MDGPS appears independent of other clinicopathological characteristics. In terms of predictive capacity and clinical usefulness, the MDGPS was superior to traditional TNM stage. Additionally, the MDGPS was confirmed in external LSCC cohorts from GEO. CMap matched the 9 most significant small molecules as promising therapeutic drugs to reverse the LSCC gene expression. Finally, CISH analysis in 30 LSCC tissues and paired adjacent normal tissues revealed that MAGEB2 has significantly higher expression of LSCC compared to adjacent non-neoplastic tissues; LINC01354, CCDC8, PHYHD1, and ZNF732 have significantly lower expression of LSCC compared to adjacent non-neoplastic tissues, which were in line with bioinformatics analysis results.

Conclusion

A MDGPS, with five DNA MDGs, was identified and validated in LSCC patients by combining transcriptome and methylation datasets analysis. Compared TNM stage alone, it generates more accurate estimations of the recurrence prediction and maybe offer novel research directions and prospects for individualized treatment of patients with LSCC.
Appendix
Available only for authorised users
Literature
1.
go back to reference Unsal AA, Kilic S, Dubal PM, Baredes S, Eloy JA. A population-based comparison of European and North American sinonasal cancer survival. Auris Nasus Larynx. 2018;45(4):815–24.CrossRef Unsal AA, Kilic S, Dubal PM, Baredes S, Eloy JA. A population-based comparison of European and North American sinonasal cancer survival. Auris Nasus Larynx. 2018;45(4):815–24.CrossRef
2.
go back to reference Siegel RL, Miller KD. Cancer statistics, 2020. Cancer J Clin. 2020;70(1):7–30.CrossRef Siegel RL, Miller KD. Cancer statistics, 2020. Cancer J Clin. 2020;70(1):7–30.CrossRef
3.
go back to reference Lo Nigro C, Denaro N, Merlotti A, Merlano M. Head and neck cancer: improving outcomes with a multidisciplinary approach. Cancer Manag Res. 2017;9:363–71.CrossRef Lo Nigro C, Denaro N, Merlotti A, Merlano M. Head and neck cancer: improving outcomes with a multidisciplinary approach. Cancer Manag Res. 2017;9:363–71.CrossRef
4.
go back to reference Cui J, Wang L, Tan G, et al. Development and validation of nomograms to accurately predict risk of recurrence for patients with laryngeal squamous cell carcinoma: Cohort study. Int J Surg. 2020;76:163–70.CrossRef Cui J, Wang L, Tan G, et al. Development and validation of nomograms to accurately predict risk of recurrence for patients with laryngeal squamous cell carcinoma: Cohort study. Int J Surg. 2020;76:163–70.CrossRef
5.
go back to reference Cui J, Wen Q, Tan X, Chen Z. A genomic-clinicopathologic nomogram predicts survival for patients with laryngeal squamous. Cell Carcinoma. 2019;2019:5980567. Cui J, Wen Q, Tan X, Chen Z. A genomic-clinicopathologic nomogram predicts survival for patients with laryngeal squamous. Cell Carcinoma. 2019;2019:5980567.
6.
go back to reference Stefansson OA, Moran S, Gomez A, Sayols S, Arribas-Jorba C, Sandoval J, Hilmarsdottir H, Olafsdottir E, Tryggvadottir L, Jonasson JG, et al. A DNA methylation-based definition of biologically distinct breast cancer subtypes. Mol Oncol. 2015;9(3):555–68.CrossRef Stefansson OA, Moran S, Gomez A, Sayols S, Arribas-Jorba C, Sandoval J, Hilmarsdottir H, Olafsdottir E, Tryggvadottir L, Jonasson JG, et al. A DNA methylation-based definition of biologically distinct breast cancer subtypes. Mol Oncol. 2015;9(3):555–68.CrossRef
7.
go back to reference Teschendorff AE, Gao Y, Jones A, Ruebner M, Beckmann MW, Wachter DL, Fasching PA, Widschwendter M. DNA methylation outliers in normal breast tissue identify field defects that are enriched in cancer. Nat Commun. 2016;7:10478.CrossRef Teschendorff AE, Gao Y, Jones A, Ruebner M, Beckmann MW, Wachter DL, Fasching PA, Widschwendter M. DNA methylation outliers in normal breast tissue identify field defects that are enriched in cancer. Nat Commun. 2016;7:10478.CrossRef
8.
go back to reference Bao M, Shi R, Zhang K, Zhao Y, Wang Y. Development of a membrane lipid metabolism-based signature to predict overall survival for personalized medicine in ccRCC patients. EPMA J. 2019;10(4):383–93.CrossRef Bao M, Shi R, Zhang K, Zhao Y, Wang Y. Development of a membrane lipid metabolism-based signature to predict overall survival for personalized medicine in ccRCC patients. EPMA J. 2019;10(4):383–93.CrossRef
9.
go back to reference Long J, Chen P, Lin J, Bai Y, Yang X, Bian J, Lin Y, Wang D, Yang X, Zheng Y, et al. DNA methylation-driven genes for constructing diagnostic, prognostic, and recurrence models for hepatocellular carcinoma. Theranostics. 2019;9(24):7251–67.CrossRef Long J, Chen P, Lin J, Bai Y, Yang X, Bian J, Lin Y, Wang D, Yang X, Zheng Y, et al. DNA methylation-driven genes for constructing diagnostic, prognostic, and recurrence models for hepatocellular carcinoma. Theranostics. 2019;9(24):7251–67.CrossRef
10.
go back to reference Vickers AJ, Van Calster B, Steyerberg EW. Net benefit approaches to the evaluation of prediction models, molecular markers, and diagnostic tests. BMJ. 2016;352:i6.CrossRef Vickers AJ, Van Calster B, Steyerberg EW. Net benefit approaches to the evaluation of prediction models, molecular markers, and diagnostic tests. BMJ. 2016;352:i6.CrossRef
11.
go back to reference Shen Z, Lin L, Cao B, Zhou C, Hao W, Ye D. LZTS2 promoter hypermethylation: a potential biomarker for the diagnosis and prognosis of laryngeal squamous cell carcinoma. World J Surg Oncol. 2018;16(1):42.CrossRef Shen Z, Lin L, Cao B, Zhou C, Hao W, Ye D. LZTS2 promoter hypermethylation: a potential biomarker for the diagnosis and prognosis of laryngeal squamous cell carcinoma. World J Surg Oncol. 2018;16(1):42.CrossRef
12.
go back to reference Wang Y, Zhang ZX, Chen S, Qiu GB, Xu ZM, Fu WN. Methylation status of SP1 Sites within miR-23a-27a-24–2 promoter region influences laryngeal cancer cell proliferation and apoptosis. Biomed Res Int. 2016;2016(206):1248. Wang Y, Zhang ZX, Chen S, Qiu GB, Xu ZM, Fu WN. Methylation status of SP1 Sites within miR-23a-27a-24–2 promoter region influences laryngeal cancer cell proliferation and apoptosis. Biomed Res Int. 2016;2016(206):1248.
13.
go back to reference Jili S, Eryong L, Lijuan L, Chao Z. RUNX3 inhibits laryngeal squamous cell carcinoma malignancy under the regulation of miR-148a-3p/DNMT1 axis. Cell Biochem Funct. 2016;34(8):597–605.CrossRef Jili S, Eryong L, Lijuan L, Chao Z. RUNX3 inhibits laryngeal squamous cell carcinoma malignancy under the regulation of miR-148a-3p/DNMT1 axis. Cell Biochem Funct. 2016;34(8):597–605.CrossRef
14.
go back to reference Qiu J, Peng B, Tang Y, et al. CpG Methylation signature predicts recurrence in early-stage hepatocellular carcinoma: results from a multicenter study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2017;35(7):734–42.CrossRef Qiu J, Peng B, Tang Y, et al. CpG Methylation signature predicts recurrence in early-stage hepatocellular carcinoma: results from a multicenter study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2017;35(7):734–42.CrossRef
15.
go back to reference Hoban CW, Beesley LJ, Bellile EL, et al. Individualized outcome prognostication for patients with laryngeal cancer. Cancer. 2018;124(4):706–16.CrossRef Hoban CW, Beesley LJ, Bellile EL, et al. Individualized outcome prognostication for patients with laryngeal cancer. Cancer. 2018;124(4):706–16.CrossRef
16.
go back to reference Te Riele RJLM, Dronkers EAC, Wieringa MH, et al. Influence of anemia and BMI on prognosis of laryngeal squamous cell carcinoma: Development of an updated prognostic model. Oral Oncol. 2018;78:25–30.CrossRef Te Riele RJLM, Dronkers EAC, Wieringa MH, et al. Influence of anemia and BMI on prognosis of laryngeal squamous cell carcinoma: Development of an updated prognostic model. Oral Oncol. 2018;78:25–30.CrossRef
17.
go back to reference Cedoz PL, Prunello M, Brennan K, Gevaert O. MethylMix 2.0: an R package for identifying DNA methylation genes. Bioinformatics. 2018;34(17):3044–6.CrossRef Cedoz PL, Prunello M, Brennan K, Gevaert O. MethylMix 2.0: an R package for identifying DNA methylation genes. Bioinformatics. 2018;34(17):3044–6.CrossRef
18.
go back to reference Berger DM, Wassenberg RM, Jozwiak K, Wiel BA, Balm JM, Berg JG, Klop WM. Inter-observer variation in the histopathology reports of head and neck melanoma; a comparison between the seventh and eighth edition of the AJCC staging system. Eur J Surg Oncol. 2019;45(2):235–241. Berger DM, Wassenberg RM, Jozwiak K, Wiel BA, Balm JM, Berg JG, Klop WM. Inter-observer variation in the histopathology reports of head and neck melanoma; a comparison between the seventh and eighth edition of the AJCC staging system. Eur J Surg Oncol. 2019;45(2):235–241.
19.
go back to reference Li J, He M, Xu W, Huang S. LINC01354 interacting with hnRNP-D contributes to the proliferation and metastasis in colorectal cancer through activating Wnt/beta-catenin signaling pathway. J Exp Clin Cancer Res. 2019;38(1):161.CrossRef Li J, He M, Xu W, Huang S. LINC01354 interacting with hnRNP-D contributes to the proliferation and metastasis in colorectal cancer through activating Wnt/beta-catenin signaling pathway. J Exp Clin Cancer Res. 2019;38(1):161.CrossRef
20.
go back to reference Lu W, Xu Y, Xu J, Wang Z, Ye G. Identification of differential expressed lncRNAs in human thyroid cancer by a genome-wide analyses. Cancer Med. 2018;7(8):3935–44.CrossRef Lu W, Xu Y, Xu J, Wang Z, Ye G. Identification of differential expressed lncRNAs in human thyroid cancer by a genome-wide analyses. Cancer Med. 2018;7(8):3935–44.CrossRef
21.
go back to reference Wang P, Yan F, Li Z, Yu Y, Parnell SE, Xiong Y. Impaired plasma membrane localization of ubiquitin ligase complex underlies 3-M syndrome development. J Clin Investig. 2019;129(10):4393–407.CrossRef Wang P, Yan F, Li Z, Yu Y, Parnell SE, Xiong Y. Impaired plasma membrane localization of ubiquitin ligase complex underlies 3-M syndrome development. J Clin Investig. 2019;129(10):4393–407.CrossRef
22.
go back to reference Pangeni RP, Channathodiyil P, Huen DS, Eagles LW, Johal BK, Pasha D, Hadjistephanou N, Nevell O, Davies CL, Adewumi AI, et al. The GALNT9, BNC1 and CCDC8 genes are frequently epigenetically dysregulated in breast tumours that metastasise to the brain. Clin Epigenet. 2015;7:57.CrossRef Pangeni RP, Channathodiyil P, Huen DS, Eagles LW, Johal BK, Pasha D, Hadjistephanou N, Nevell O, Davies CL, Adewumi AI, et al. The GALNT9, BNC1 and CCDC8 genes are frequently epigenetically dysregulated in breast tumours that metastasise to the brain. Clin Epigenet. 2015;7:57.CrossRef
23.
go back to reference Furusawa Y, Kubo T, Fukazawa T. Phyhd1, an XPhyH-like homologue, is induced in mouse T cells upon T cell stimulation. Biochem Biophys Res Commun. 2016;472(3):551–6.CrossRef Furusawa Y, Kubo T, Fukazawa T. Phyhd1, an XPhyH-like homologue, is induced in mouse T cells upon T cell stimulation. Biochem Biophys Res Commun. 2016;472(3):551–6.CrossRef
24.
go back to reference Jin S, Cao S, Li J, et al. Cancer/testis antigens (CTAs) expression in resected lung cancer. OncoTargets Therapy. 2018;11:4491–9.CrossRef Jin S, Cao S, Li J, et al. Cancer/testis antigens (CTAs) expression in resected lung cancer. OncoTargets Therapy. 2018;11:4491–9.CrossRef
25.
go back to reference Nobeyama Y, Nakagawa H. Aberrant demethylation and expression of MAGEB2 in a subset of malignant peripheral nerve sheath tumors from neurofibromatosis type 1. J Dermatol Sci. 2016;81(2):118–23.CrossRef Nobeyama Y, Nakagawa H. Aberrant demethylation and expression of MAGEB2 in a subset of malignant peripheral nerve sheath tumors from neurofibromatosis type 1. J Dermatol Sci. 2016;81(2):118–23.CrossRef
26.
go back to reference Van Tongelen A, Loriot A, De Smet C. Oncogenic roles of DNA hypomethylation through the activation of cancer-germline genes. Cancer Lett. 2017;396:130–7.CrossRef Van Tongelen A, Loriot A, De Smet C. Oncogenic roles of DNA hypomethylation through the activation of cancer-germline genes. Cancer Lett. 2017;396:130–7.CrossRef
27.
go back to reference Pattani KM, Soudry E, Glazer CA, Ochs MF, Wang H, Schussel J, Sun W, Hennessey P, Mydlarz W, Loyo M, et al. MAGEB2 is activated by promoter demethylation in head and neck squamous cell carcinoma. PLoS ONE. 2012;7(9):e45534.CrossRef Pattani KM, Soudry E, Glazer CA, Ochs MF, Wang H, Schussel J, Sun W, Hennessey P, Mydlarz W, Loyo M, et al. MAGEB2 is activated by promoter demethylation in head and neck squamous cell carcinoma. PLoS ONE. 2012;7(9):e45534.CrossRef
28.
go back to reference Reuter S, Prasad S, Phromnoi K, Ravindran J, Sung B, Yadav VR, Kannappan R, Chaturvedi MM, Aggarwal BB. Thiocolchicoside exhibits anticancer effects through downregulation of NF-kappaB pathway and its regulated gene products linked to inflammation and cancer. Cancer Prev Res. 2010;3(11):1462–72.CrossRef Reuter S, Prasad S, Phromnoi K, Ravindran J, Sung B, Yadav VR, Kannappan R, Chaturvedi MM, Aggarwal BB. Thiocolchicoside exhibits anticancer effects through downregulation of NF-kappaB pathway and its regulated gene products linked to inflammation and cancer. Cancer Prev Res. 2010;3(11):1462–72.CrossRef
Metadata
Title
Identification and validation of methylation-driven genes prognostic signature for recurrence of laryngeal squamous cell carcinoma by integrated bioinformatics analysis
Authors
Jie Cui
Liping Wang
Waisheng Zhong
Zhen Chen
Jie Chen
Hong Yang
Genglong Liu
Publication date
01-12-2020
Publisher
BioMed Central
Keyword
Epigenetics
Published in
Cancer Cell International / Issue 1/2020
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-020-01567-3

Other articles of this Issue 1/2020

Cancer Cell International 1/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine