Identification of prognostic gene biomarkers for metastatic skin cancer using data mining

  • Authors:
    • Gang Liu
    • Chen Li
    • Haiyan Zhen
    • Zhigang Zhang
    • Yongzhong Sha
  • View Affiliations

  • Published online on: May 18, 2020     https://doi.org/10.3892/br.2020.1307
  • Pages: 22-30
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Skin cancer is a common malignant tumor in China and throughout the world, and the rate of recurrence is considerably high, thus endangering the quality of life and health of patients, and increasing the economic burden and pressure to the families of those afflicted. Due to the limitations of traditional drug treatments, it is difficult to achieve the desired therapeutic effect of complete removal. However, targeted gene therapy may be a novel means of treating skin cancer, as the targeted nature of treatment may improve therapeutic outcomes. However, targeted gene therapy requires physicians to select the appropriate gene, which means suitable genetic biomarkers must be identified from complex genetic data. In the present study, the least absolute shrinkage and selection operator regression analysis method was used with 10‑fold cross verification to reduce the dimensions of gene data in patients with skin cancer, and subsequently, 20 gene biomarkers were screened. A prognostic model was constructed using these 20 gene biomarkers, and the validity of the model was assessed using a training set and a verification set, which showed that the model performed well. Finally, gene function analysis of these 20 gene biomarkers was determined. Relevant studies were found to show that the genetic biomarkers identified in this paper may possess value for the follow‑up clinical treatment of skin cancer.

Introduction

Skin cancer is a malignant tumor that afflicts individuals all over the world. Skin cancer is divided into malignant melanoma and non-melanoma skin cancer (NMSC) (1,2). According to global cancer statistics in 2018, there were 142,056 new cases of NMSC, accounting for 5.8% of global cancer cases, and 65,155 NMSC-related deaths, accounting for 0.7% of global cancer mortality. There were 779,723 new cases of skin malignant melanoma, accounting for 1.6% of global cancer cases, and 60,712 skin melanoma-related deaths, accounting for 0.6% of global cancer mortality (3).

According to the 2015 Chinese Cancer Statistics Report, the incidence of skin cancer in China was 8/1,000 and the mortality rate was 3.22/1,000 (4,5). Compared with other types of cancer, skin cancer has a higher recurrence rate, with a 35% probability of recurrence in the first 3 years and a 50% probability of recurrence in the first 5 years. Recurrent skin cancer is usually the same sub-type as the original cancer (6). Although the mortality rate of skin cancer is extremely low and the cure rate is high, its high incidence and recurrence rate constitute a significant economic burden to health services. Furthermore, skin cancer located on the head and other highly visible areas may affect a patients mental wellbeing and quality of life (7,8).

Cutaneous metastases of malignant tumors can be caused by malignant tumor cells traveling through the blood or lymphatic system, tissue interstitial diffusion (tissue gap diffusion), or surgical implantation (9). The higher the malignancy of the tumor, the more likely it is to metastasize, and skin metastasis is often the end-stage manifestation of a malignant tumor (10). Once metastasis occurs, the prognosis of cancer is poor (11). Unfortunately, skin metastasis may be the first clinical manifestation of a malignant tumor (12). Therefore, a suitable prognosis serves an important role in the recurrence and treatment of metastatic skin cancer. The aim of the present study was to identify potential prognostic biomarkers of metastatic skin cancer, which may be used in a clinical setting, through data mining and analysis of skin cancer prognosis genes.

With the development of gene chip technology and next-generation sequencing technology (13), and the constant revision of the viewpoints of individualized medical treatment and precision medical care, understanding skin cancer from the perspective of the genome and proposing more effective genetic biomarkers provides more relevant information for drug development and clinical decision-making (14-18). Traditional statistical methods often yield unstable results and excessive errors when applied to gene expression analysis (19). Furthermore, in high-throughput gene expression experiments, the number of variables is generally much higher than the sample size, which is called the Curse of Dimensionality (20). Therefore, in the present study, least absolute shrinkage and selection operator (LASSO) was used to mine genomic data to minimize the instability caused by high-dimensional data (21-27).

Materials and methods

Data collection

Using The Cancer Genome Atlas, data on skin cancer from the Xena Functional Genomics Explorer (xenabrowser.net/datapages/) was obtained, including the number of patients (n=481) and the number of genes assessed (n=20,530). Among these, the number of primary samples (Primary Tumor) was 105, the number of metastatic (Metastatic) samples was 368, and the number of other types of samples was 8 (28-32).

Data preprocessing

First, 219 samples with missing survival attribute values were removed from 473 primary and metastatic samples and 255 patient samples were retained. These data were stratified into primary tumor samples (n=72) and metastatic samples (n=183) by matching the patients' samples in the gene expression spectrum. Finally, using the ‘sample’ function in R version 3.5.2(33), the metastatic samples were randomly divided into two groups; training samples (n=91) and test samples (n=92).

Clinical data analysis

Several clinical factors affect the prognosis of patients with skin cancer, so it is necessary for data analysis to consider multiple attribute values in the sample as influencing factors. Based on previous studies, clinical variables including age, sex, history of radiotherapy, Tumor-Node-Metastasis pathological stage (34) and cancer status were used to analyze the related clinical factors in the subsequent data mining analysis (35-38).

Analysis of gene data

First, the ‘Limma’ package version 3.42.2(39) in R was used to analyze the differentially expressed genes of the 91 training set samples and 72 primary tumor samples. According to the adjusted P-value (adj.P.val<0.001), 783 genes were considered to be differentially expressed genes.

The ‘survival’ package (rdocumentation.org/packages/survival; version 3.1-8) in R was used to perform Cox regression analysis of the differentially expressed genes and the Cox coefficient, the hazard ratio (HR) and the P-values of the Wald test of each gene were calculated using the Kaplan-Meier method (40), and the genes that were significantly associated with the survival of the patients (P<0.05) were screened using the ‘survivalROC’ package (version number 1.0.3; rdocumentation.org/packages/survivalROC).

Finally, the screened genes were analyzed again using LASSO in the R package ‘glmnet’ (rdocumentation.org/packages/glmnet; version 3.0-2), to obtain more critical genes. Through 10-fold cross validation, 20 risk genes, which were closely related to survival, were identified (41-49). Additionally, the Gene Ontology (GO) Cell Component Ontology Method (50,51) was used to analyze pathway involved in protein activity.

Prognostic index (PI) calculation

As an important indicator of the integration of risk genes, a PI value can be determined for each patient with skin cancer. The PI was obtained by linearly fitting the product of the expression and the coefficient corrected by LASSO of each gene. The formula for calculating the PI was: PI=β1 X12X2+…+βiXi…+βnXn; where Xi is the expression of the ith gene and βi is the coefficient corrected by LASSO of the ith gene.

Data validation

By extrapolating genes with a P<0.05, the 20 risk genes obtained were verified using the test sample using the same methods described above.

Statistical analysis of clinical variables

From the clinical information of 255 patient samples, the patients' age, sex, radiation therapy, pathological T-stage and cancer status were taken as single variables. Cox regression analysis was performed, and the P-value of the Log-rank test and the HR value of each clinical factor were calculated. P<0.05 was considered to indicate a statistically significant difference.

Results

Clinicopathological variables

Age, pathologic T-stage and cancer status were all significantly associated with the recurrence of skin cancer, suggesting that these three clinical factors may be used as independent prognostic indicators (Table I).

Table I

Clinicopathological characteristics of the patients.

Table I

Clinicopathological characteristics of the patients.

CharacteristicsNumber of patients (relapse)P-valueHR (95% CI)
Age 0.0301.669 (1.037-2.688)
     >57123(35)  
     ≤57132(36)  
Sex 0.2001.359 (0.8058-2.291)
     Male160(49)  
     Female95(22)  
Cancer status   
     Unknown3(1) 7x10-10 
     With tumor160(18) 0.802 (0.1089-5.908)
     Tumor free92(52) 0.1572 (0.0206-1.201)
Radiation therapy 0.2001.7204 (0.851-3.478)
     Unknown1 (0)  
     Yes38(9)  
     No216(62)  
Pathological T-stage 0.0092.003 (1.175-3.412)
     T1-T3165(49)  
     T490(22)  

[i] HR, hazard ratio; CI, confidence interval.

Genetic data analysis

After determining differential gene expression, 44 genes were considered significantly associated with the survival of patients using survival analysis. Cox regression analysis and LASSO analysis were performed, and 20 risk genes associated with skin cancer were identified (Table II).

Table II

Prognostic genes.

Table II

Prognostic genes.

Gene symbolNameUnivariate HR (95% CI)CoefficientP-valueLASSO HR (95% CI)
TMEM45BTransmembrane protein 45B0.899 (0.822-0.982)-0.086 1.88x10-20.918 (0.864-0.975)
CDKN1BCyclin-dependent kinase inhibitor 1B (p27, Kip1)0.928 (0.886-0.972)-0.029 1.55x10-30.971 (0.914-1.032)
PKHD1L1Polycystic kidney and hepatic disease 1 (autosomal recessive)-like 10.938 (0.886-0.993)-0.010 2.73x10-20.990 (0.932-1.052)
PLCL2Phospholipase C-like 20.961 (0.929-0.994)-0.006 2.14x10-20.994 (0.936-1.056)
CKMT1ACreatine kinase, mitochondrial 1A1.024 (1.000-1.049)0.001 4.90x10-21.001 (0.942-1.063)
SPINK5Serine peptidase inhibitor, Kazal type 51.027 (1.002-1.052)0.007 3.31x10-21.008 (0.948-1.070)
CST6Cystatin E/M1.027 (1.000-1.054)0.011 4.67x10-21.011 (0.952-1.074)
FABP5Fatty acid binding protein 5 (psoriasis-associated)1.037 (1.007-1.067)0.007 1.53x10-21.007 (0.948-1.070)
PTK6Protein tyrosine kinase 61.047 (1.010-1.084)0.029 1.11x10-21.030 (0.969-1.094)
TXNDC17Thioredoxin domain containing 171.047 (1.002-1.095)0.004 4.02x10-21.004 (0.945-1.066)
LTB4RLeukotriene B4 receptor1.050 (1.007-1.096)0.018 2.39x10-21.018 (0.958-1.082)
FAM100AFamily with sequence similarity 100, member A1.050 (1.008-1.094)0.015 1.90x10-21.016 (0.956-1.079)
KRT10keratin 101.057 (1.025-1.091)0.016 4.33x10-41.016 (0.956-1.080)
PPP1R13LProtein phosphatase 1, regulatory subunit 13 like1.059 (1.010-1.109)0.000 1.65x10-21.000 (0.941-1.063)
DLX3Distal-less homeobox 31.063 (1.025-1.102)0.008 9.40x10-41.008 (0.949-1.071)
KPRPKeratinocyte proline rich protein1.064 (1.015-1.115)0.044 9.68x10-31.045 (0.984-1.111)
VSIG10LV-set and immunoglobulin domain containing 10 like1.066 (1.011-1.123)0.021 1.87x10-21.021 (0.961-1.085)
MYOM3Myomesin 31.073 (1.015-1.134)0.040 1.25x10-21.041 (0.979-1.106)
NMUNeuromedin U receptor 11.092 (1.042-1.144)0.072 2.22x10-41.075 (1.012-1.142)
PIGW Phosphatidylinositol glycan anchor biosynthesis, class W1.113 (1.044-1.188)0.002 1.08x10-31.002 (0.943-1.064)

[i] HR, hazard ratio; CI, confidence interval.

Gene PI analysis

Through linear fitting of the product of expression and regression coefficient of the 20 genes in each sample, the PI of each patient was calculated, and the patients were sorted from lowest to highest according to their PI value. Based on the median PI value, the patients were divided into high-risk and low-risk groups (Fig. 1). The lower the PI value, the lower the risk of recurrent survival, and the higher the PI value, the higher the risk of recurrent survival.

Using the Kaplan Meier method, the survival curves for the two groups of patients, are presented in Fig. 2A; patients considered low risk exhibited significantly longer overall survival times (P<0.001; HR=26.321). Using the 5-year survival rates, a Receiver Operating Characteristic (ROC) curve was drawn (Fig. 2B) and analysis was performed using the ‘survivalROC’ package. The advantages and disadvantages of the model constructed using the 20 gene biomarkers were determined based on the Area Under the Curve (AUC) value. The results showed that AUC was equal to 0.908 (AUC >0.5 indicates a suitable model). The heat map of risk gene expression profiles in high-risk and low-risk patients were plotted (Fig. 3). The high-risk group was clearly distinguished from the low-risk group. This indicated that the models constructed by these 20 gene biomarkers performed well.

Based on the experimental results, the 20 gene prognostic biomarkers could be used to significantly classify the patients with skin cancer in the training samples into two groups: High risk and low risk. In order to further verify the accuracy of the test results, the 20 genetic biomarkers were used to validate the test samples. As shown in Fig. 4, these genetic biomarkers could still classify patients with skin cancer in the test sample into high-risk and low-risk categories (P=0.004, HR=1.194). The AUC of the ROC curve was 0.855.

Prognostic gene function analysis

The online genetic analysis tool STRING (52) was used to analyze and study the association between the identified gene biomarkers and their associated protein synthesis pathways (Fig. 5; Table III) Gene Ontology (GO) cellular component ontology analysis identified a pathway of which involved a cyclin-dependent protein kinase holoenzyme complex, which included three proteins (CCND1, CDK2 and CDK4).

Table III

GO biological processes associated with the prognostic genes.

Table III

GO biological processes associated with the prognostic genes.

Pathway IDPathway descriptionGene countFalse discovery rateMatching proteins
GO.0010948Negative regulation of cell cycle process50.0219CCNA2, CCND1, CDK2, CDK4, CDKN1B
GO.0031100Organ regeneration40.0219CCNA2, CCND1, CDK2, CDK4
GO.0044773mitotic DNA Damage checkpoint40.0219CCNA2, CCND1, CDK2, CDKN1B
GO.0071156Regulation of cell cycle arrest40.0219CCND1, CDK2, CDK4, CDKN1B
GO.1901990Regulation of mitotic cell cycle phase transition50.0219CCNA2, CCND1, CDK2, CDK4, CDKN1B
GO.0032355Response to estradiol40.0238CCNA2, CCND1, CDK2, CDKN1B
GO.0010389Regulation of G2/M transition of mitotic cell cycle30.0277CCNA2, CCND1, CDK4
GO.0044772Mitotic cell cycle phase transition50.0277CCNA2, CCND1, CDK2, CDK4, CDKN1B
GO.0097305Response to alcohol50.0277CCNA2, CCND1, CDK2, CDK4, CDKN1B
GO.1901991Negative regulation of mitotic cell cycle phase transition40.0287CCNA2, CCND1, CDK2, CDKN1B
GO.0000082G1/S transition of mitotic cell cycle40.0326CCND1, CDK2, CDK4, CDKN1B
GO.0048513Organ development110.0345CCNA2, CCND1, CDK2, CDK4, CDKN1B, DLX3, KRT1, KRT10, PLCL2, PPP1R13L, SPINK5
GO.0060429Epithelium development70.0457CCND1, CST6, DLX3, FABP5, KRT10, PTK6, SPINK5
GO.0048545Response to steroid hormone50.0482CCNA2, CCND1, CDK2, CDK4, CDKN1B

[i] GO, Gene Ontology.

Performance of the biomarkers in clinical subtypes

Among the clinical factors, pathological T stage (T1 and T2-4), cancer status (with tumor and tumor free) and age (>57 years vs. ≤57 years) were all significantly associated with the recurrence status of patients with skin cancer (Fig. 6). Therefore, these 20 genes should to be considered in different clinical types to determine which clinical state they are more suitable for.

The results in Fig. 6 shows the predictive effect of these 20 gene biomarkers. Patients who were tumor free had improved survival compared with those with tumor. Similarly, patients with a T-stage of T1-3 exhibited improved survival compares with patients classed as T4, and patients ≤57 years old had improved survival compared with patients >57 years old.

Discussion

In the present study, a variety of statistical analysis methods were used (including LASSO regression, single-factor survival analysis, multi-factor Cox proportional hazards regression model and ROC curve analysis) to identify differences in the gene expression profiles of patients with skin cancer. A supervised cluster analysis method was used and 20 prognostic genes from the training samples were identified and verified against a test sample. The results showed that these 20 genes can stratify patients with skin cancer as high-risk and low-risk, which shows the feasibility of the mining method used in the present study.

From a biological point of view, the 20 prognostic genes identified in the present study successfully divided the patients with cancer according to the risk of recurrence, which may have important reference value for the treatment of recurrence of metastatic skin cancer, and may influence future clinical studies of skin cancer and drug development.

Among the 20 prognostic genes identified in the present study, several are closely associated with skin cancer and metastatic skin cancer, including DLX3, PTK6 and CST6 genes. For example, physical interaction between DLX3 and P53 on P21 promoter can enhance the expression of P21(53). Increasing DLX3 expression in keratinocytes produces a G1-S blockade associated with P53 signature transcriptional profiles (54).

Deletion of DLX3 promotes a mitogenic phenotype associated with constitutive activation of ERK (55). The loss of DLX3 expression in human skin cancer suggests that a DLX3-P53 interaction may be a primary regulatory axis of epidermis differentiation and that DLX3 may be a regulator of skin cancer development. Protein tyrosine kinase 6 (PTK6) is expressed in ~70% of cases of triple-negative breast cancer, in which it serves an important role in promoting metastatic lung colonization and survival (56). PTK6 inhibits the inhibition of metastasis of triple-negative breast cancer via SNAIL-dependent regulation of E-cadherin expression (57). Epigenetic changes associated with upregulation of CST6 gene expression may be accompanied by metastatic diffusion of primary tumor sites, and current studies have shown that methylation-dependent epigenetic silencing of CST6 represents an important mechanism for loss of CST6 during the development and/or progression to metastasis (58). Other gene prognostic biomarkers identified in the present study have potential research value and need further exploration and research.

Among these, several genes, including PLCL2, serine protease inhibitor Kazal type-5 (SPINK5) and KRT10 are associated with skin diseases. Inosine strongly enhances proliferation of human C32 melanoma cells through the PLCL2 and PI3K pathways (59). SPINK5 serves a crucial role in the timing of desquamation of the skin (60). Biallelic KRT10 mutations result in skin fragility caused by self-improving epidermolytic ichthyosis (61). Several other genes, such as transmembrane protein 45B (TMEM45B), CDKN1B, CKMT1A, FABP5 and PPP1R13L have also been shown to be associated with several types of cancer. TMEM45B has been shown to be abnormally expressed in gastric tumors and serves an important role in gastric tumorigenesis (62). The CCND1-A870G and CDKN1B-C79T polymorphisms are associated with breast cancer risk (63). The n335586/miR-924/CKMT1A axis contributes to migration and invasion of hepatocellular carcinoma cells (64). FABP5 serves an important role in the carcinogenesis and metastasis of cervical cancer, and may be a novel predictor for prognostic assessment of patients with cervical cancer (65). Polymorphisms of ERCC1, CD3EAP and PPP1R13L in chromosomal region 19q13.2-3 have previously been shown to exhibit a synergistic effect on apoptosis and DNA repair pathways (66).

In summary, the 20 gene biomarkers identified based on the LASSO algorithm can effectively predict the risk of patients with skin cancer and may be more convenient as a model of prognosis.

Acknowledgements

Not applicable.

Funding

This work was supported by the Key Project of China Ministry of Education for Philosophy and Social Science: Big Data Driven Risk Research on City's Public Safety (grant no. 16JZD023) and the Fundamental Research Funds for the Central Universities: Big Data Driven Risk Pre-Warning Research on City's Public Safety (grant no. 17LZUJBWZD012).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

GL analyzed and interpreted the patient data was a major contributor in writing the manuscript. CL analyzed the data. HZ made substantial contributions to acquisition of data. ZZ was a major contributor in interpreting the data and in writing the manuscript. YS made substantial contributions to analysis and interpretation of data. All authors have 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 

Diepgen TL and Mahler V: The epidemiology of skin cancer. Br J Dermatol. 146 (Suppl 61):S1–S6. 2002.PubMed/NCBI View Article : Google Scholar

2 

Lai V, Cranwell W and Sinclair R: Epidemiology of skin cancer in the mature patient. Clin Dermatol. 36:167–176. 2018.PubMed/NCBI View Article : Google Scholar

3 

Pilgrim W, Hayes R, Hanson DW, Zhang B, Boudreau B and Leonfellner S: Skin Cancer (Basal Cell Carcinoma, Squamous Cell Carcinoma, and Malignant Melanoma): New cases, treatment practice, and health care costs in New Brunswick, Canada, 2002-2010. J Cutan Med Surg. 18:320–331. 2014.PubMed/NCBI View Article : Google Scholar

4 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in china, 2015. CA Cancer J Clin. 66:115–132. 2016.PubMed/NCBI View Article : Google Scholar

5 

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.PubMed/NCBI View Article : Google Scholar

6 

Karagas MR, Stukel TA, Greenberg ER, Baron JA, Mott LA and Stern RS: Risk of subsequent basal cell carcinoma and squamous cell carcinoma of the skin among patients with prior skin cancer. Skin cancer prevention study group. JAMA. 267:3305–3310. 1992.PubMed/NCBI

7 

Jerant AF, Johnson JT, Sheridan CD and Caffrey TJ: Early detection and treatment of skin cancer. Am Fam Physician. 62:357–368, 375-376, 381-382. 2000.PubMed/NCBI

8 

Gauci J and Muscat G: A local perspective on basal cell carcinoma: Frequency of subsequent skin tumours. Malta Med J. 1:46–55. 2017.

9 

Gillner J, Kirchberg K, Korge B, Hunzelmann N, Krieg T and Scharffetter-Kochanek K: Cutaneous metastases from a leiomyosarcoma of the testicular tunica albuginea. Hautarzt. 51:41–45. 2000.PubMed/NCBI View Article : Google Scholar : (In German).

10 

Hoyt BS and Cohen PR: Cutaneous scrotal metastasis: Origins and clinical characteristics of visceral malignancies that metastasize to the scrotum. Int J Dermatol. 52:398–405. 2013.PubMed/NCBI View Article : Google Scholar

11 

Bremnes RM, Veve R, Hirsch FR and Franklin WA: The E-cadherin cell-cell adhesion complex and lung cancer invasion, metastasis, and prognosis. Lung Cancer. 36:115–124. 2002.PubMed/NCBI View Article : Google Scholar

12 

Zhen Y, Wu C and Jiang J: Analysis of clinicopathological features and prognostic factors in cutaneous metastases of malignant tumors. Chin Clin Oncol. 19:152–155. 2014.

13 

Niu JX, Meng XK and Ren JJ: Studied microRNA gene expression in human hepatocellular carcinoma by microRNA microarray techniques. World J Gastroenterol. 21:12605–12611. 2015.PubMed/NCBI View Article : Google Scholar

14 

Hou X, Peng JX, Hao XY, Cai JP, Liang LJ, Zhai JM, Zhang KS, Lai JM and Yin XY: DNA methylation profiling identifies EYA4 gene as a prognostic molecular marker in hepatocellular carcinoma. Ann Surg Oncol. 21:3891–3899. 2014.PubMed/NCBI View Article : Google Scholar

15 

Pivarcsi A and Sonkoly E: Skin cancer associated microRNAs. US Patent 12/994,734. Filed June 4, 2009; issued May 5, 2011.

16 

Glavač D and Ravnik-Glavač M: Essential role of microRNA in skin physiology and disease. Adv Exp Med Biol. 888:307–330. 2015.PubMed/NCBI View Article : Google Scholar

17 

Leffell DJ: The scientific basis of skin cancer. J Am Acad Dermatol. 42 (Suppl):S18–S22. 2000.PubMed/NCBI View Article : Google Scholar

18 

Thyagarajan A, Shaban A and Sahu RP: MicroRNA-directed cancer therapies: Implications in melanoma intervention. J Pharmacol Exp Ther. 364:1–12. 2018.PubMed/NCBI View Article : Google Scholar

19 

Liu Z, Chen D, Xu Y and Liu J: Logistic support vector machines and their application to gene expression data. Int J Bioinform Res Appl. 1:169–182. 2005.PubMed/NCBI View Article : Google Scholar

20 

Yongchao GE, Sealfon SC and Speed TP: Multiple testing and its applications to microarrays. Stat Methods Med Res. 18:543–563. 2009.PubMed/NCBI View Article : Google Scholar

21 

Okimoto G, Zeinalzadeh A, Wenska T, Loomis M, Nation JB, Fabre T, Tiirikainen M, Hernandez B, Chan O, Wong L and Kwee S: Joint analysis of multiple high-dimensional data types using sparse matrix approximations of rank-1 with applications to ovarian and liver cancer. BioData Min. 9(24)2016.PubMed/NCBI View Article : Google Scholar

22 

Xiong J, Xiong K and Bing Z: Clinical and RNA expression integrated signature for urothelial bladder cancer prognosis. Cancer Biomark. 21:535–546. 2018.PubMed/NCBI View Article : Google Scholar

23 

Tang RX, Chen WJ, He RQ, Zeng JH, Liang L, Li SK, Ma J, Luo DZ and Chen G: Identification of a RNA-Seq based prognostic signature with five lncRNAs for lung squamous cell carcinoma. Oncotarget. 8:50761–50773. 2017.PubMed/NCBI View Article : Google Scholar

24 

Gao X, Wu Y, Yu W and Li H: Identification of a seven-miRNA signature as prognostic biomarker for lung squamous cell carcinoma. Oncotarget. 7:81670–81679. 2016.PubMed/NCBI View Article : Google Scholar

25 

van Malenstein H, Gevaert O, Libbrecht L, Daemen A, Allemeersch J, Nevens F, Van Cutsem E, Cassiman D, De Moor B, Verslype C and van Pelt J: A seven-gene set associated with chronic hypoxia of prognostic importance in hepatocellular carcinoma. Clin Cancer Res. 16:4278–4288. 2010.PubMed/NCBI View Article : Google Scholar

26 

Nakazawa H, English D, Randell PL, Nakazawa K, Martel N, Armstrong BK and Yamasaki H: UV and skin cancer: Specific p53 gene mutation in normal skin as a biologically relevant exposure measurement. Proc Natl Acad Sci USA. 91:360–364. 1994.PubMed/NCBI View Article : Google Scholar

27 

Linck L, Liebig J, Völler D, Eichner N, Lehmann G, Meister G and Bosserhoff A: MicroRNA-sequencing data analyzing melanoma development and progression. Exp Mol Pathol. 105:371–379. 2018.PubMed/NCBI View Article : Google Scholar

28 

Goldman M, Craft B, Swatloski T, Ellrott K, Cline M, Diekhans M, Ma S, Wilks C, Stuart J, Haussler D and Zhu J: The UCSC Cancer Genomics Browser: Update 2013. Nucleic Acids Res. 41 (Database Issue):D949–D954. 2013.PubMed/NCBI View Article : Google Scholar

29 

Cline MS, Craft B, Swatloski T, Goldman M, Ma S, Haussler D and Zhu J: Exploring TCGA Pan-Cancer Data at the UCSC Cancer Genomics Browser. Sci Rep. 3(2652)2013.PubMed/NCBI View Article : Google Scholar

30 

Goldman M, Craft B, Swatloski T, Cline M, Morozova O, Diekhans M, Haussler D and Zhu J: The UCSC cancer genomics browser: Update 2015. Nucleic Acids Res. 43 (Database Issue):D812–D817. 2015.PubMed/NCBI View Article : Google Scholar

31 

Turner N, Ware O and Bosenberg M: Genetics of metastasis: Melanoma and other cancers. Clin Exp Metastasis. 35:379–391. 2018.PubMed/NCBI View Article : Google Scholar

32 

Kong J, Zhang Y and Zhao B: A clinicopathological analysis of 104 cases with metastatic tumors of the skin. Chin J Diagnostic Pathol. 2:211–213. 1995.

33 

R Core Team: R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, 2006.

34 

Loh KC, Greenspan FS, Gee L, Miller TR and Yeo PP: Pathological Tumor-Node-Metastasis (pTNM) staging for papillary and follicular thyroid carcinomas: A retrospective analysis of 700 patients. J Clin Endocrinol Metab. 82:3553–3562. 1997.PubMed/NCBI View Article : Google Scholar

35 

Lezcano C, Shoushtari AN, Ariyan C, Hollmann TJ and Busam KJ: Primary and metastatic melanoma with NTRK fusions. Am J Surg Pathol. 42:1052–1058. 2018.PubMed/NCBI View Article : Google Scholar

36 

Rhee JS, Matthews BA, Neuburg M, Logan BR, Burzynski M and Nattinger AB: The skin cancer index: Clinical responsiveness and predictors of quality of life. Laryngoscope. 117:399–405. 2007.PubMed/NCBI View Article : Google Scholar

37 

Morgan M, McCreedy R, Simpson J and Hay RJ: Dermatology quality of life scales: A measure of the impact of skin diseases. Br J Dermatol. 136:202–206. 1997.PubMed/NCBI

38 

Rogers EM, Connolly KL, Nehal KS, Dusza SW, Rossi AM and Lee E: Comorbidity scores associated with limited life expectancy in the very elderly with nonmelanoma skin cancer. J Am Acad Dermatol. 78:1119–1124. 2018.PubMed/NCBI View Article : Google Scholar

39 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43(e47)2015.PubMed/NCBI View Article : Google Scholar

40 

Nagy A, Lánczky A, Menyhárt O and Győrffy B: Validation of miRNA prognostic power in hepatocellular carcinoma using expression data of independent datasets. Sci Rep. 8(9227)2018.PubMed/NCBI View Article : Google Scholar

41 

Zaorsky NG, Lee CT, Zhang E and Galloway TJ: Skin CanceR Brachytherapy vs External beam radiation therapy (SCRiBE) meta-analysis. Radiother Oncol. 126:386–393. 2018.PubMed/NCBI View Article : Google Scholar

42 

Riker AI, Enkemann SA, Fodstad O, Liu S, Ren S, Morris C, Xi Y, Howell P, Metge B, Samant RS, et al: The gene expression profiles of primary and metastatic melanoma yields a transition point of tumor progression and metastasis. BMC Med Genomics. 1(13)2008.PubMed/NCBI View Article : Google Scholar

43 

Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, Carter SL, Stewart C, Mermel CH, Roberts SA, et al: Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 499:214–218. 2013.PubMed/NCBI View Article : Google Scholar

44 

Wiltgen M, Gerger A and Smolle J: Tissue counter analysis of benign common nevi and malignant melanoma. Int J Med Inform. 69:17–28. 2003.PubMed/NCBI View Article : Google Scholar

45 

Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012.PubMed/NCBI View Article : Google Scholar

46 

Esteva A, Kuprel B, Novoa RA, Ko J, Swetter SM, Blau HM and Thrun S: Dermatologist-level classification of skin cancer with deep neural networks. Nature. 542:115–118. 2017.PubMed/NCBI View Article : Google Scholar

47 

Esteva A, Kuprel B, Novoa RA, Ko J, Swetter SM, Blau HM and Thrun S: Corrigendum: Dermatologist-level classification of skin cancer with deep neural networks. Nature. 546(686)2017.PubMed/NCBI View Article : Google Scholar

48 

Masood A and Al-Jumaily AA: Computer aided diagnostic support system for skin cancer: A review of techniques and algorithms. Int J Biomed Imaging. 2013(323268)2013.PubMed/NCBI View Article : Google Scholar

49 

Chang P, Bing Z, Tian J, Zhang J, Li X, Ge L, Ling J, Yang K and Li Y: Comprehensive assessment gene signatures for clear cell renal cell carcinoma prognosis. Medicine (Baltimore). 97(e12679)2018.PubMed/NCBI View Article : Google Scholar

50 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: Tool for the unification of biology The Gene Ontology Consortium. Nat Genet. 25:25–29. 2000.PubMed/NCBI View Article : Google Scholar

51 

The Gene Ontology Consortium. The Gene Ontology Resource: 20 years and still GOing strong. Nucleic Acids Res. 47:D330–D338. 2019.PubMed/NCBI View Article : Google Scholar

52 

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.PubMed/NCBI View Article : Google Scholar

53 

Panno ML, Giordano F, Mastroianni F, Morelli C, Brunelli E, Palma MG, Pellegrino M, Aquila S, Miglietta A, Mauro L, et al: Evidence that low doses of Taxol enhance the functional transactivatory properties of p53 on p21 waf promoter in MCF-7 breast cancer cells. FEBS Lett. 580:2371–2380. 2006.PubMed/NCBI View Article : Google Scholar

54 

Palazzo E, Kellett MD, Cataisson C, Bible PW, Bhattacharya S, Sun HW, Gormley AC, Yuspa SH and Morasso MI: A novel DLX3-PKC integrated signaling network drives keratinocyte differentiation. Cell Death Differ. 24:717–730. 2017.PubMed/NCBI View Article : Google Scholar

55 

Jiang L, Campagne C, Sundström E, Sousa P, Imran S, Seltenhammer M, Pielberg G, Olsson MJ, Egidy G, Andersson L and Golovko A: Constitutive activation of the ERK pathway in melanoma and skin melanocytes in Grey horses. BMC Cancer. 14(857)2014.PubMed/NCBI View Article : Google Scholar

56 

Palazzo E, Kellett M, Cataisson C, Gormley A, Bible PW, Pietroni V, Radoja N, Hwang J, Blumenberg M, Yuspa SH and Morasso MI: The homeoprotein DLX3 and tumor suppressor p53 co-regulate cell cycle progression and squamous tumor growth. Oncogene. 35:3114–3124. 2016.PubMed/NCBI View Article : Google Scholar

57 

Ito K, Park SH, Nayak A, Byerly JH and Irie HY: PTK6 Inhibition suppresses metastases of triple-negative breast cancer via SNAIL-dependent E-cadherin regulation. Cancer Res. 76:4406–4417. 2016.PubMed/NCBI View Article : Google Scholar

58 

Rivenbark AG, Livasy CA, Boyd CE, Keppler D and Coleman WB: Methylation-dependent silencing of CST6 in primary human breast tumors and metastatic lesions. Exp Mol Pathol. 83:188–197. 2007.PubMed/NCBI View Article : Google Scholar

59 

Soares AS, Costa VM, Diniz C and Fresco P: Inosine strongly enhances proliferation of human C32 melanoma cells through PLC-PKC-MEK1/2-ERK1/2 and PI3K pathways. Basic Clin Pharmacol Toxicol. 116:25–36. 2015.PubMed/NCBI View Article : Google Scholar

60 

Le NA, Katsuyama M, Demura M, Tanii H, Katsuyama H and Saijoh K: Regulation of serine protease inhibitor Kazal type-5 (SPINK5) gene expression in the keratinocytes. Environ Health Prev Med. 19:307–313. 2014.PubMed/NCBI View Article : Google Scholar

61 

Frommherz L, Küsel J, Zimmer A, Fischer J and Has C: Skin fragility caused by biallelic KRT10 mutations: An intriguing form of self-improving epidermolytic ichthyosis. Br J Dermatol. 182:780–785. 2020.PubMed/NCBI View Article : Google Scholar

62 

Shen K, Yu W, Yu Y, Liu X and Cui X: Knockdown of TMEM45B inhibits cell proliferation and invasion in gastric cancer. Biomed Pharmacother. 104:576–581. 2018.PubMed/NCBI View Article : Google Scholar

63 

Canbay E, Eraltan IY, Cercel A, Isbir T, Gazioglu E, Aydogan F, Cacina C, Cengiz A, Ferahman M, Zengin E and Unal H: CCND1 and CDKN1B polymorphisms and risk of breast cancer. Anticancer Res. 30:3093–3098. 2010.PubMed/NCBI

64 

Fan H, Lv P, Mu T, Zhao X, Liu Y, Feng Y, Lv J, Liu M and Tang H: LncRNA n335586/miR-924/CKMT1A axis contributes to cell migration and invasion in hepatocellular carcinoma cells. Cancer Lett. 429:89–99. 2018.PubMed/NCBI View Article : Google Scholar

65 

Wang W, Chu HJ, Liang YC, Huang JM, Shang CL, Tan H, Liu D, Zhao YH, Liu TY and Yao SZ: FABP5 correlates with poor prognosis and promotes tumor cell growth and metastasis in cervical cancer. Tumor Biol. 37:14873–14883. 2016.PubMed/NCBI View Article : Google Scholar

66 

Chae YS, Kim JG, Kang BW, Lee SJ, Jeon HS, Park JS, Choi GS and Lee WK: PPP1R13Lvariant associated with prognosis for patients with rectal cancer. J Cancer Res Clin Oncol. 139:465–473. 2013.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

July-2020
Volume 13 Issue 1

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu G, Li C, Zhen H, Zhang Z and Sha Y: Identification of prognostic gene biomarkers for metastatic skin cancer using data mining. Biomed Rep 13: 22-30, 2020
APA
Liu, G., Li, C., Zhen, H., Zhang, Z., & Sha, Y. (2020). Identification of prognostic gene biomarkers for metastatic skin cancer using data mining. Biomedical Reports, 13, 22-30. https://doi.org/10.3892/br.2020.1307
MLA
Liu, G., Li, C., Zhen, H., Zhang, Z., Sha, Y."Identification of prognostic gene biomarkers for metastatic skin cancer using data mining". Biomedical Reports 13.1 (2020): 22-30.
Chicago
Liu, G., Li, C., Zhen, H., Zhang, Z., Sha, Y."Identification of prognostic gene biomarkers for metastatic skin cancer using data mining". Biomedical Reports 13, no. 1 (2020): 22-30. https://doi.org/10.3892/br.2020.1307