Identification of a multidimensional transcriptome signature predicting tumor regrowth of clinically non‑functioning pituitary adenoma

  • Authors:
    • Sen Cheng
    • Jing Guo
    • Yazhuo Zhang
    • Zhenye Li
    • Chuzhong Li
  • View Affiliations

  • Published online on: June 23, 2020     https://doi.org/10.3892/ijo.2020.5087
  • Pages: 804-812
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Clinically non‑functioning pituitary adenoma (NFPA) represents approximately one third of all pituitary adenomas. Tumor regrowth is an important feature of NFPA; however, the effective methods with which to predict this are limited. The present study analyzed the expression of protein‑coding genes and long non‑coding RNA in 66 patients with NFPA. Cox regression analysis was performed to identify genes associated with regrowth or progression‑free survival (PFS). Kaplan‑Meier, random survival forest analysis and receiver operating characteristic curve (ROC) analyses were performed to generate a multi‑protein‑coding gene (PCG) and long non‑coding RNA (lncRNA) signature with a maximum area under the ROC curve (AUC). In total, 1 PCG (CHST12) and 2 lncRNAs (COA6‑AS1 and RP11‑23N2.4) were identified that were significantly associated with tumor regrowth. The multi‑transcriptome signature exhibited a high predictive accuracy for tumor regrowth, with an AUC of 0.869/0.726 in the training/testing set, and the discriminative power of this signature was better than that of age. On the whole, the present study indicates that the combined PCG and lncRNA signature may be beneficial as a marker for the prediction of the prognosis of patients with NFPA.

Introduction

Clinically non-functioning pituitary adenoma (NFPA) is one of the most common subtypes of pituitary adenoma. Being unassociated with specific serum hormone changes, NFPA is usually detected based on symptoms, such as headaches and visual disturbance or incidental imaging examinations (1,2). Surgical resection is the primary treatment choice for NFPA; however, patients are often faced with tumor residue as the tumor may have surrounded the internal carotid artery or invaded the cavernous sinus (3,4). Approximately 12-58% of patients may experience relapse within 5 years (5). Radiotherapy is often recommended to patients with tumor residue; however, its long-term complications, such as visual defects and hypopituitarism, pose concerns (6,7). Thus, surgery remains the optimal treatment option for patients with tumor regrowth. However, in contrast to clinically functioning pituitary adenoma (FPA), for which serum hormone monitoring is used for detection, NFPA lacks an effective evaluation method, resulting in the failure of early intervention. Thus, research concerning the molecular mechanisms of tumor regrowth and the development of methods for predicting prognosis is of utmost significance.

Protein-coding genes (PCGs) are the most commonly used class of molecular marker. Numerous studies have reported that the altered expression of PCGs results in aggressive tumor behavior and may affect the prognosis of patients with NFPA (1,8,9). Long non-coding RNAs (lncRNAs) are a subgroup of non-protein-coding RNAs >200 nucleotides in length. lncRNAs play pivotal roles in the regulation of gene expression and have been used as prognostic markers in multiple tumors types (8,10). The expression of lncRNAs has been found to be related to the growth and invasive behavior of pituitary adenoma (11,12). However, there has been little research on the role of lncRNAs or its combination with PCGs as potentially novel approaches for prognostic evaluation in pituitary adenoma.

The present study aimed to identify PCGs and lncRNAs that are asscoiated with the regrowth of NFPA, and to establish a model which may be used to predict tumor regrowth based on the PCG and lncRNA expression profiles of 66 NFPA samples through microarray analyses. The findings of the present study may provide an effective method for the prediction of the prognosis and post-operative intervention of patients with NFPA.

Materials and methods

Patients and samples

NFPA specimens from 66 patients who had undergone surgical treatment at Beijing Tiantan Hospital from October, 2007 to July, 2014 were selected, which included 34 females and 32 males with a median age of 55 years (range, 25-66 years). The median follow-up time was 78 months (range, 36-121 months). The definition of tumor regrowth was a maximum increase in tumor diameter of >2 mm on an enhanced MRI from the time of surgery to the follow-up endpoint with or without headaches, visual disturbances or other mass effect symptoms. The clinical characteristics of the patients are presented in Table I and detailed clinical information is presented in Table SI. The present study was approved by the Medical Ethics Committee of Beijing Tiantan Hospital and written informed consent for the use of the resected samples for research purposes was obtained from all patients.

Table I

Clinical characteristics of the 66 patients with NFPA.

Table I

Clinical characteristics of the 66 patients with NFPA.

FeatureTraining setTesting setEntire set
Sex
 Female151934
 Male181432
Age (years)
 ≤52181533
 >52151833
Knosp classification
 I5914
 II549
 III5510
 IV181533
Regrowth
 Yes232346
 No101020
Invasion
 Yes211836
 No121530

[i] NFPA, non-functioning pituitary adenoma.

Total RNA extraction and RNA microarray

Total cellular RNA was extracted from the tumor samples using the mirVana™ miRNA Isolation kit without phenol (cat. no. AM1561, Ambion; Thermo Fisher Scientific, Inc.), which were used to produce labeled fluorescent cRNA targets for the SBC human ceRNA array v1.0 (4x180 K). The prepared cRNA targets were hybridized with the slides and scanned on an Agilent Microarray Scanner (Agilent Technologies, Inc.). Following data collection with Feature Extraction software v10.7 (Agilent Technologies, Inc.), the data were normalized by quantile normalization using a package named limma from R program (bioinf.wehi.edu.au/limma).

Selection of PCGs and lncRNAs subsets related to tumor regrowth

All patients We were randomly divided into the training (n=33) and testing sets (n=33) using an algorithm called sample from R program (www.r-project.org/). In the training set, the evaluation of the association between the expression of candidate PCGs and lncRNAs, and the PFS of each patient was performed using univariate Cox regression analysis.

Selection of candidate PCGs and lncRNAs as a predictive signature

A model for selecting PCGs and lncRNAs as a predictive signature of prognosis was developed using the following formula (13,14):

Risk score(RS)=∑Ni=1(Explg×Coef), where 'N' in the formula represents the number of prognostic PCGs and lncRNAs, 'Explg' represents the expression of PCGs and lncRNAs, and 'Coef' represents the regression coefficient of PCGs and lncRNAs.

Random survival forests-variable hunting (RSFVH) analysis was used to screen 10 PCGs and lncRNAs, as previously described (13). Considering that a prediction model with a smaller number of PCGs and lncRNAs would be more practical than one with a larger number, all of the screened PCGs and lncRNAs were included. A time-dependent ROC curve was used to evaluate the specificity and sensitivity of the regrowth prediction of the RS of the 210-1=1023 signatures in the training set. The patients with NFPA in each set were separated into the high- and low-risk groups with the median RS in the training set used as the cut-off value.

Statistical analysis

Kaplan-Meier survival analyses were performed to assess the regrowth distributions in the training and testing sets, and the statistical significance was assessed using the two-sided log-rank test. Chi-square tests were performed to analyze the associations among the clinical features. Multivariable Cox regression analysis was performed to evaluate the independence of the risk score from clinical features. P<0.05 was considered to indicate a statistically significant in the present study. All the analyses in the present study were performed in the R program (www.r-project.org) with the timeROC (cran.r-project.org/web/packages/timeROC/index.html), survivalROC (cran.r-project.org/web/packages/survivalROC/index.html) and randomForestSRC packages (cran.r-project.org/package=randomForestSRC).

Functional analysis of PCGs and lncRNAs

The co-expression association between the candidate PCGs and lncRNAs was evaluated with Pearson's correlation coefficients. The expression data used for Pearson's correlation were derived from the microarray analysis. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analyses of the PCGs and lncRNAs were performed to confirm the biological functions of lncRNAs. The co-expression correlations between the candidate PCGs and all other PCGs were assessed using Pearson's correlation analysis, and genes with P-values <0.05 and absolute values from the Pearson's correlation coefficient >0.6 were selected for KEGG and GO enrichment analyses. The above-mentioned analyses were performed with the clusterProfiler package (15,16).

Results

Construction of a PCG-lncRNA signature for the prediction of NFPA regrowth

Expression profiles of 66 pituitary tumor tissues were obtained using a ceRNA microarray. The expression levels of 18,829 PCGs and 19,741 lncRNAs were then determined. The 66 NFPAs were randomly divided into a training set (n=33) and a testing set (n=33). The training set was used for screening the candidate PCGs and lncRNAs associated with tumor regrowth, and the testing set was used for validating the classification power. In the selection step, all analyses were based on the training set.

With regrowth as the dependent variable, univariate Cox regression analysis was performed. In total, 1,245 PCGs and 1,214 lncRNAs (Table SII) were identified that were signifi-cantly associated with tumor regrowth (P<0.05; Fig. 1A and B). Subsequently, 10 candidates, including 5 PCGs and 5 lncRNAs related to tumor regrowth were identified among the 2,459 members of the PCG and lncRNA set according to the permutation importance values of the random forest supervised classification (RSFC) analysis (Fig. 1C and D).

To obtain an optimized prediction signature, time-dependent ROC analysis was performed and the specificity and sensitivity of the regrowth prediction of the RS was evaluated for each patient according to the RS model of 210-1=1023 in the training set (Table SIII). The PCG and lncRNA combination with the max AUC was composed of CHST12, COA6-AS1 and RP11-23N2.4 (Fig. 1C and D; Table II). The RS of the signature was obtained as follows: RS = (4.95 x CHST12) + (3.41 x COA 6-AS1) + (1.90 x RP11-23N2.4). The AUC for the PCG and lncRNA signature was 0.869 in the training set, which indicated a strong performance for regrowth prediction (Fig. 1E and F).

Table II

Identification of PCGs and lncRNAs in the predicting signature and the univariable Cox association with regrowth.

Table II

Identification of PCGs and lncRNAs in the predicting signature and the univariable Cox association with regrowth.

Gene symbolCoefficientaP-valueaExpression level association with poor prognosisChromosome location (GRCh38/hg38)
CHST124.950.003Highchr7: 2403588-2448484: +
COA6-AS13.410.001High chr1:234372807-234373593: −
RP11-23N2.41.900.007High chr15:52584907-52587652: +

a Derived from the univariable Cox regression analysis in the training set. PCG, protein-coding gene; lncRNA, long non-coding RNA.

Predictive ability of the molecular signature for tumor regrowth

In the training set, patients were separated into the high-risk (n=16) and low-risk (n=17) group using the median RS of the signature as the cut-off. The results revealed that patients in the high-risk group had a shorter PFS than patients in the low-risk group (median PFS, 4.44 years vs. <7.97 years; P<0.001; Fig. 2A). The regrowth rate in the high-risk group was >50%, whereas that of the low-risk group was <1%. In the testing set, patients were similarly separated into the high-risk (n=16) and low-risk (n=17) groups. To confirm the predictive power of the CHST12, COA6-AS1 and RP11-23N2.4 signature, the results of Kaplan-Meier analysis for the patients in the testing set were plotted, as shown in Fig. 2B (median PFS, 5.51 years vs. <8.30 years; P=0.021). The regrowth rate of the patients in the low-risk group was approximately 12.6%, whereas that of patients in the high-risk group was 55.0%.

The regrowth status of patients with NFPA from the training and testing sets is illustrated in the dot plots (Fig. 2C and D). Moreover, the expression patterns of the 3 PCG and lncRNAs in the 2 groups are presented. Consistent with the pattern observed in the training set, patients with low-risk scores tended to have lower expression levels of the 1 PCG and 2 lncRNAs than the patients in the high-risk group.

Associations between patient clinical characteristics and the molecular signature

To better understand the associations between clinical characteristics and the CHST12, COA6-AS1 and RP11-23N2.4 signature, the associations of the signature with the clinical characteristics of the whole patient set (n=66) were investigated. Unlike age, sex, and invasion status based on Knosp classification (17,18) exhibited no association with the PCG-lncRNA signature (Table III).

Table III

Association of the signature with the clinical characteristics of the patients with NFPA.

Table III

Association of the signature with the clinical characteristics of the patients with NFPA.

VariableTraining set
P-valueaTesting set
P-valueaEntire set
P-valuea
Low riskHigh riskLow riskHigh riskLow riskHigh risk
Sex> 0.990.620.63
 Female871181915
 Male99681517
Age (years)0.590.020.03
 ≤528104111221
 >52961352211
Invasion> 0.990.390.48
 Yes11101172217
 No66691215

a Data were analyzed using the Chi-squared test; P-value <0.05 was considered to indicate a statistically significant difference. NFPA, non-functioning pituitary adenoma.

To explore the independency of our signature from the clinical characteristics, multivariable Cox regression analysis was performed of the entire patient set using the PCG-lncRNA signature-based RS. It was found that the CHST12, COA6-AS1 and RP11-23N2.4 signature was indeed independent of these clinical characteristics (high-risk vs. low-risk, HR=1.47; 95% CI, 1.22-1.79; P<0.001, n=66; Table IV).

Table IV

Univariate and multivariate Cox regression analysis of the signature of the patients with NFPA in the training, testing set and entire set.

Table IV

Univariate and multivariate Cox regression analysis of the signature of the patients with NFPA in the training, testing set and entire set.

A, Univariable analysis
VariableHRTraining set (n=33) 95% CI
P-valueHRTesting set (n=33) 95% CI
P-valueHREntire set (n=66) 95% CI
P-value
LowerUpperLowerUpperLowerUpper
Sex
 Male vs. female1.050.293.740.940.530.142.030.350.750.311.810.52
Age (years)
 >52 vs. ≤520.230.051.090.060.350.091.360.130.290.100.790.02
Knosp classification
 III IV vs. I II1.360.722.540.341.110.681.830.671.210.821.780.33
Signature
 High-risk vs. low-risk1.881.372.57<0.0011.240.921.660.041.471.231.76<0.001

B, Multivariable analysis

VariableHRTraining set (n=33) 95% CI
P-valueHRTesting set (n=33) 95% CI
P-valueHREntire set (n=66) 95% CI
P-value
LowerUpperLowerUpperLowerUpper

Sex
 Male vs. female0.540.093.400.520.490.122.030.330.500.171.450.20
Age (years)
 >52 vs. ≤520.290.051.820.190.540.132.240.390.470.181.230.12
Knosp classification
 III IV vs. I II1.390.662.920.381.140.661.960.641.200.821.750.35
Signature
 High-risk vs. low-risk1.951.342.86<0.0011.230.891.700.051.471.221.79<0.001

[i] NFPA, non-functioning pituitary adenoma.

Comparison of the predictive power between the molecular signature and age

To compare the specificity and sensitivity of regrowth prediction between age and our signature, ROC analysis was performed. The predictive power of our signature was significantly better than that of age in the training/testing set (AUC=0.870/0.683 vs. AUC=0.726/0.676; Fig. 3A and B), which demonstrated that our signature is an effective for prediction.

To further understand the predictive ability of the signature regarding 3- to 5-year PFS, timeROC analysis was used within the 2 sets. The AUC of our signature was 0.767/0.818/0.833 at 3/4/5 years in the training set and 0.651/0.607/0.713 at 3/4/5 years in the testing set, respectively (Fig. 3C and D). The results indicated that our predictive signature had a better performance for 5-year PFS than for 3- or 4-year PFS.

Functional characterization of the PCG and lncRNAs composing the prognostic signature

To explore the biological functions of the PCG and lncRNAs in our signature, the co-expression network of the one PCG and two lncRNAs with other PCGs were assessed using Pearson's correlation analysis. The expression levels of 1,076 PCGs highly correlated with those of one or more PCG and lncRNAs (Pearson's correlation coefficient >0.60, P<0.05, Table SIV). Subsequently, performed KEGG pathway and GO function enrichment analyses were performed for the co-expressed PCGs. GO function annotation analysis revealed that 1,074 PCGs were enriched in 98 GO/KEGG terms (Table SV), which indicated that the PCG and lncRNAs may be associated with important biological processes, including those involving the spliceosome and mitochondrial protein complex, etc., (Fig. 4).

Discussion

Unlike tumor regrowth in functioning pituitary adenoma, which can be monitored by monitoring the levels of specific serum hormones, tumor regrowth in NFPA is difficult to monitor. By the time patients are re-examined due to optic nerve compression symptoms, the tumor may have acquired a large volume, which presents a number of obstacles to total resection and post-operative recovery. Therefore, the present study aimed to develop novel predictive signatures that can identify early regrowth and serve as predictive markers of prognosis. The present study aimed to categorize patients into a high- and low-risk group; thus the most effective and timely treatment can be applied to patients with NFPA.

A previous study focused on factors associated with tumor regrowth in NFPA with the ultimate goal of ameliorating the prognosis of patients at the post-operative stage (18). Age is recognized as an important independent factor influencing the prognosis of patients with NFPA, with a younger age being associated with a greater chance of tumor regrowth (19,20). However, the prognostic value of age is not as effective as the PCG-lncRNA signature identified in the present study. Ki-67 is another commonly used pathological prognostic evaluation index (21); however, the use of a single indicator for prognostic assessment has limitations in accurately evaluating patient prognosis. A previous study attempted to establish a statistical model that considers both combines clinical features (age and tumor volume) and molecular markers (p16, WIF1 and TGF-β) to evaluate regrowth probability among patients with NFPA post-operatively (1). In the present study, the inclusion of clinical characteristics did not improve predictive efficacy. Furthermore, unlike the above-mentioned study, a time component was added to the prognosis assessment and independently assessed the prognosis of patients at different time points.

In recent years, long non-coding RNAs have been reported in a number of types of tumor and represent a promising novel group of molecular markers for tumor biological behavior, disease diagnosis and prognosis evaluation (22-24). The expression of lncRNA H19 has been found to be decreased in pituitary adenomas, and its overexpression can markedly inhibit the growth of pituitary tumor cells; furthermore, it has been identified as a potential drug resistance marker (11). Xing et al (25), identified differentially expressed mRNAs and lncRNAs in clinically NFPA and normal pituitary, and constructed a mRNA-lncRNA co-expression network. However, their research failed to illustrate the regulatory mechanisms of key genes or lncRNAs or their effects on patient prognosis. Guo et al (26) successfully constructed a circRNA signature using the random survival forest algorithm, Kaplan-Meier analysis and ROC analysis. They randomly divided patients into a training and testing set and obtained an accuracy of 0.87 and 0.67 in each group. Compared with the single signature, the present study included PCGs and lncRNAs into the model and achieved a better accuracy in this model. In addition, the present study introduced a concept of time into our predictive signature, which could be more instructive in clinical practice. The present study found 2 lncRNAs that could be used as prognostic signatures. However, the function and regulatory mechanisms of these two lncRNA have not yet been reported. A PCG marker, CHST12, was also identified, which plays an important role in articular cartilage; however, there is limited research available on its role in tumors (27,28). The biological roles of the one PCG and two lncRNAs composing the signature in the present study are not yet fully illustrated and warrant further investigation in future studies.

There are a few limitations to the present study which need to be acknowledged. First, there is limited research available on the functions and molecular mechanisms of these PCGs and lncRNAs in tumors. The biological roles of CHST12, COA6-AS1 and RP11-23N2.4 have not yet been fully illustrated and thus require further investigation in the future. Second, there are limited sequencing data available regarding NFPA; thus, the verification of the present results with an independent validating set was not possible. In addition, further studies are required using larger cohort sizes and for the validation of the present results in the future. It should be noted that further research based on these data is still ongoing and the authors would like to share their data privately to researchers who are interested in pituitary adenoma. Finally, the application of our signature in clinical practice should be tested prospectively. Despite the above-mentioned limitations, the consistent and significant correlation of our CHST12, COA6-AS1 and RP11-23N2.4 signature with tumor regrowth indicates that this signature is a potentially potent prognostic predictive model for NFPA.

In conclusion, to the best of our knowledge, this is the first study to integrate PCGs and lncRNAs to predict tumor regrowth in patients with NFPA. The findings of the present study may provide a new aspect of prognostic evaluation and may help patients to benefit from early intervention.

Supplementary Data

Acknowledgments

The authors would like to thank Dr Yazhuo Miao and Mr. Qiuyue Fang from Capital Medical University for the selfless assistance in collecting clinical data.

Funding

The present study was funded by the National Natural Science Foundation of China (grant nos. 81771489, 81672495 and 81601205), the National High Technology Research and Development Program of China (863 Program, grant no. 2014AA020610), the Beijing Municipal Science and Technology Commission (grant no. Z171100000117002), the Beijing Talents Fund (grant no. 2015000021223ZK24) and the China National Key Research and Development Program (grant no. 2017YFC0908300).

Availability of data and materials

Further research regarding the topic of the present study is ongoing and all data in this study are available from the corresponding author on reasonable request.

Authors' contributions

SC was involved in data collection and analysis, and in the writing of the manuscript. JG and ZL were involved in data collection. CL and YZ were involved in the conception and design of the research, funding management and in the final editing of the manuscript.

Ethics approval and consent to participate

The present study was approved by the Medical Ethics Committee of Beijing Tiantan Hospital and written informed consent for the use of the resected samples for research purposes was obtained from all patients.

Patient consent for publication

Not applicable.

Competing interests

All authors declare that they have no competing interests.

References

1 

Cheng S, Wu J, Li C, Li Y, Liu C, Li G, Li W, Hu S, Ying X and Zhang Y: Predicting the regrowth of clinically non-functioning pituitary adenoma with a statistical model. J Transl Med. 17:1642019. View Article : Google Scholar : PubMed/NCBI

2 

Ebersold MJ, Quast LM, Laws ER Jr, Scheithauer B and Randall RV: Long-term results in transsphenoidal removal of nonfunctioning pituitary adenomas. J Neurosurg. 64:713–719. 1986. View Article : Google Scholar : PubMed/NCBI

3 

Meij BP, Lopes MB, Ellegala DB, Alden TD and Laws ER Jr: The long-term significance of microscopic dural invasion in 354 patients with pituitary adenomas treated with transsphenoidal surgery. J Neurosurg. 96:195–208. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Shomali ME and Katznelson L: Medical therapy of gonadotropin-producing and nonfunctioning pituitary adenomas. Pituitary. 5:89–98. 2002. View Article : Google Scholar

5 

Brochier S, Galland F, Kujas M, Parker F, Gaillard S, Raftopoulos C, Young J, Alexopoulou O, Maiter D and Chanson P: Factors predicting relapse of nonfunctioning pituitary macroadenomas after neurosurgery: A study of 142 patients. Eur J Endocrinol. 163:193–200. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Brada M and Jankowska P: Radiotherapy for pituitary adenomas. Endocrinol Metab Clin North Am. 37:pp. 263–275. pp. xi2008, https://doi.org/10.1016/j.ecl.2007.10.005. View Article : Google Scholar

7 

Pollock BE, Cochran J, Natt N, Brown PD, Erickson D, Link MJ, Garces YI, Foote RL, Stafford SL and Schomberg PJ: Gamma knife radiosurgery for patients with nonfunctioning pituitary adenomas: Results from a 15-year experience. Int J Radiat Oncol Biol Phys. 70:1325–1329. 2008. View Article : Google Scholar

8 

Aydin B and Arga KY: Co-expression network analysis elucidated a core module in association with prognosis of non-functioning non-invasive human pituitary adenoma. Front Endocrinol (Lausanne). 10:3612019. View Article : Google Scholar

9 

Zhenye L, Chuzhong L, Youtu W, Xiaolei L, Lei C, Lichuan H, Hongyun W, Yonggang W, Fei W and Yazhuo Z: The expression of TGF-β1, Smad3, phospho-Smad3 and Smad7 is correlated with the development and invasion of nonfunctioning pituitary adenomas. J Transl Med. 12:712014. View Article : Google Scholar

10 

Dykes IM and Emanueli C: Transcriptional and post-transcriptional gene regulation by long non-coding RNA. Genomics Proteomics Bioinformatics. 15:177–186. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Zhang Y, Liu YT, Tang H, Xie WQ, Yao H, Gu WT, Zheng YZ, Shang HB, Wang Y, Wei YX, et al: Exosome-transmitted lncRNA H19 inhibits the growth of pituitary adenoma. J Clin Endocrinol Metab. 104:6345–6356. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Zhu H, Guo J, Shen Y, Dong W, Gao H, Miao Y, Li C and Zhang Y: Functions and mechanisms of tumor necrosis factor-α and noncoding RNAs in bone-invasive pituitary adenomas. Clin Cancer Res. 24:5757–5766. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Guo JC, Li CQ, Wang QY, Zhao JM, Ding JY, Li EM and Xu LY: Protein-coding genes combined with long non-coding RNAs predict prognosis in esophageal squamous cell carcinoma patients as a novel clinical multi-dimensional signature. Mol Biosyst. 12:3467–3477. 2016. View Article : Google Scholar : PubMed/NCBI

14 

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:e472015. View Article : Google Scholar : PubMed/NCBI

15 

Yu G, Wang LG, Han Y and He QY: clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 16:284–287. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Zhang H, Deng T, Ge S, Liu Y, Bai M, Zhu K, Fan Q, Li J, Ning T, Tian F, et al: Exosome circRNA secreted from adipocytes promotes the growth of hepatocellular carcinoma by targeting deubiquitination-related USP7. Oncogene. 38:2844–2859. 2019. View Article : Google Scholar :

17 

Knosp E, Steiner E, Kitz K and Matula C: Pituitary adenomas with invasion of the cavernous sinus space: A magnetic resonance imaging classification compared with surgical findings. Neurosurgery. 33:610–617; discussion 617-618. 1993.PubMed/NCBI

18 

Lee EH, Kim KH, Kwon JH, Kim HD and Kim YZ: Results of immunohistochemical staining of cell-cycle regulators: The prediction of recurrence of functioning pituitary adenoma. World Neurosurg. 81:563–575. 2014. View Article : Google Scholar

19 

Tampourlou M, Ntali G, Ahmed S, Arlt W, Ayuk J, Byrne JV, Chavda S, Cudlip S, Gittoes N, Grossman A, et al: Outcome of nonfunctioning pituitary adenomas that regrow after primary treatment: A study from two large UK centers. J Clin Endocrinol Metab. 102:1889–1897. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Xie Y, Burcu M, Linn DE, Qiu Y and Baer MR: Pim-1 kinase protects P-glycoprotein from degradation and enables its glycosylation and cell surface expression. Mol Pharmacol. 78:310–318. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Hasanov R, Aydoğan BI, Kiremitçi S, Erden E and Güllü S: The prognostic roles of the Ki-67 proliferation index, P53 expression, mitotic index, and radiological tumor invasion in pituitary adenomas. Endocr Pathol. 30:49–55. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Chen X, Dai M, Zhu H, Li J, Huang Z, Liu X, Huang Y, Chen J and Dai S: Evaluation on the diagnostic and prognostic values of long non-coding RNA BLACAT1 in common types of human cancer. Mol Cancer. 16:1602017. View Article : Google Scholar : PubMed/NCBI

23 

Wu ZR, Yan L, Liu YT, Cao L, Guo YH, Zhang Y, Yao H, Cai L, Shang HB, Rui WW, et al: Inhibition of mTORC1 by lncRNA H19 via disrupting 4E-BP1/Raptor interaction in pituitary tumours. Nat Commun. 9:46242018. View Article : Google Scholar : PubMed/NCBI

24 

Yamoah K, Johnson MH, Choeurng V, Faisal FA, Yousefi K, Haddad Z, Ross AE, Alshalafa M, Den R, Lal P, et al: Novel biomarker signature that may predict aggressive disease in African American men with prostate cancer. J Clin Oncol. 33:2789–2796. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Xing W, Qi Z, Huang C, Zhang N, Zhang W, Li Y, Qiu M, Fang Q and Hui G: Genome-wide identification of lncRNAs and mRNAs differentially expressed in non-functioning pituitary adenoma and construction of an lncRNA-mRNA co-expression network. Biol Open. 8:82019. View Article : Google Scholar

26 

Guo J, Wang Z, Miao Y, Shen Y, Li M, Gong L, Wang H, He Y, Gao H, Liu Q, et al: A two-circRNA signature predicts tumour recurrence in clinical non-functioning pituitary adenoma. Oncol Rep. 41:113–124. 2019.

27 

Guo Y, Zhou Y, Yan S, Qu C, Wang L, Guo X and Han J: Decreased expression of CHST-12, CHST-13, and UST in the proximal interphalangeal joint cartilage of school-age children with Kashin-Beck disease: An endemic osteoarthritis in China caused by selenium deficiency. Biol Trace Elem Res. 191:276–285. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Han J, Li D, Qu C, Wang D, Wang L, Guo X and Lammi MJ: Altered expression of chondroitin sulfate structure modifying sulfotransferases in the articular cartilage from adult osteoarthritis and Kashin-Beck disease. Osteoarthritis Cartilage. 25:1372–1375. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2020
Volume 57 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cheng S, Guo J, Zhang Y, Li Z and Li C: Identification of a multidimensional transcriptome signature predicting tumor regrowth of clinically non‑functioning pituitary adenoma. Int J Oncol 57: 804-812, 2020
APA
Cheng, S., Guo, J., Zhang, Y., Li, Z., & Li, C. (2020). Identification of a multidimensional transcriptome signature predicting tumor regrowth of clinically non‑functioning pituitary adenoma. International Journal of Oncology, 57, 804-812. https://doi.org/10.3892/ijo.2020.5087
MLA
Cheng, S., Guo, J., Zhang, Y., Li, Z., Li, C."Identification of a multidimensional transcriptome signature predicting tumor regrowth of clinically non‑functioning pituitary adenoma". International Journal of Oncology 57.3 (2020): 804-812.
Chicago
Cheng, S., Guo, J., Zhang, Y., Li, Z., Li, C."Identification of a multidimensional transcriptome signature predicting tumor regrowth of clinically non‑functioning pituitary adenoma". International Journal of Oncology 57, no. 3 (2020): 804-812. https://doi.org/10.3892/ijo.2020.5087