Next Article in Journal
Differences in Pulmonary Function Improvement after Once-Daily LABA/LAMA Fixed-Dose Combinations in Patients with COPD
Next Article in Special Issue
Development and External Validation of the STRATified CANcer Surveillance (STRATCANS) Multivariable Model for Predicting Progression in Men with Newly Diagnosed Prostate Cancer Starting Active Surveillance
Previous Article in Journal
Remarkable Remission Rate and Long-Term Efficacy of Upfront Metronomic Chemotherapy in Elderly and Frail Patients, with Diffuse Large B-Cell Lymphoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Development of a Gleason Score-Related Gene Signature for Predicting the Prognosis of Prostate Cancer

1
Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510060, China
2
Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510060, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2022, 11(23), 7164; https://doi.org/10.3390/jcm11237164
Submission received: 31 October 2022 / Revised: 24 November 2022 / Accepted: 28 November 2022 / Published: 1 December 2022
(This article belongs to the Special Issue Clinical Advances in Prostate Cancer Treatments)

Abstract

:
The recurrence of prostate cancer (PCa) is intrinsically linked to increased mortality. The goal of this study was to develop an efficient and reliable prognosis prediction signature for PCa patients. The training cohort was acquired from The Cancer Genome Atlas (TCGA) dataset, while the validation cohort was obtained from the Gene Expression Omnibus (GEO) dataset (GSE70769). To explore the Gleason score (GS)-based prediction signature, we screened the differentially expressed genes (DEGs) between low- and high-GS groups, and then univariate Cox regression survival analysis and multiple Cox analyses were performed sequentially using the training cohort. The testing cohort was used to evaluate and validate the prognostic model’s effectiveness, accuracy, and clinical practicability. In addition, the correlation analyses between the risk score and clinical features, as well as immune infiltration, were performed. We constructed and optimized a valid and credible model for predicting the prognosis of PCa recurrence using four GS-associated genes (SFRP4, FEV, COL1A1, SULF1). Furthermore, ROC and Kaplan–Meier analysis revealed a higher predictive efficiency for biochemical recurrence (BCR). The results showed that the risk model was an independent prognostic factor. Moreover, the risk score was associated with clinical features and immune infiltration. Finally, the risk model was validated in a testing cohort. Our data support that the GS-based four-gene signature acts as a novel signature for predicting BCR in PCa patients.

1. Introduction

Prostate cancer (PCa) is believed to become the second most common type of malignancy in men [1,2]. Measuring prostate-specific antigen (PSA) in the blood is a very sensitive way to detect an increased risk of lethal disease at a very early stage. The proportion of PCa is increasing rapidly each year in China. According to cancer statistics for 2020, 115,426 individuals were diagnosed with PCa and 51,095 succumbed to the malady in China [3], a level that has nearly doubled since 2015 [4]. For patients with localized curative PCa, therapeutic options such as radical prostatectomy, external beam radiotherapy, and low-dose brachytherapy are primarily recommended. The standard treatment for metastatic disease is androgen deprivation therapy [5]. The condition of an increased serum PSA level following radical prostatectomy (RP) or radiation treatment for localized PCa is known as biochemical recurrence (BCR) or biochemical relapse [6]. BCR signifies a higher risk of metastases and PCa that is resistant to castration [7]. BCR is characterized by two or even more consecutively elevated serum PSA levels of more than 0.2 ng/mL for patients who underwent radical prostatectomy [8]. Within 5 years of receiving initial therapy, approximately 20 to 30 percent of patients with clinically localized cancer will experience a clinical recurrence [9,10]. Clinical recurrence and metastasis are more likely to occur in BCR patients, especially early BCR patients, while the median survival for patients presenting with metastatic PCa is only 30 months [11]. Therefore, it is essential to discover more accurate biomarkers for the better prediction of BCR and monitoring the disease.
Tumor biomarkers used in combination with existing clinical predictors of recurrence such as Gleason score, stage, and preoperative PSA level [12,13] have been shown to provide additional information for accurately differentiating aggressive and indolent PCa patients [14]. With the advent of gene chips and high-throughput sequencing, it has become a fact that gene signatures formed from aberrant transcriptional patterns may predict prognosis for PCa patients. Multiple gene signatures for metastatic PCa prognosis prediction have been proposed in the last few years. In the case of PCa with heterogeneous hypoxia, Yang et al. proposed a 28-gene hypoxia-related signature as a powerful predictor of 5-year BCR-free survival (BFS) in PCa patients after RP or receiving post-prostatectomy radiotherapy [15]. Several studies [16] used similar methods to generate mRNA expression signatures with varying numbers of genes. Meanwhile, some aberrant gene expression information has been verified in subsequent studies, providing critical information for clinical treatment decisions [17,18]. There is thus a need to identify novel signatures that can distinguish patients at high risk for BCR.
Here, we aim to investigate a novel signature based on public data from The Cancer Genome Atlas (TCGA) database. TCGA has generated a huge amount of cancer data using high-throughput sequencing techniques and is supervised by the National Cancer Institute and the National Human Genome Research Institute [19]. The GS is the sum of the grades of the first and second Gleason patterns of a primary cancer sample. It has been clearly shown that GS have been associated with BCR [20] and prostate cancer mortality [21]. Considering GS is one of the best predictors of PCa prognosis, the strategy of finding differentially expressed transcripts in tumors stratified by GS is anticipated to acquire pertinent data on the potential for tumor aggressiveness [22]. In this study, we constructed a GS-based four-gene signature and evaluated its ability to predict recurrence after radical prostatectomy in PCa patient cohorts.

2. Materials and Methods

2.1. Gene Expression Profile and Clinical Data

Gene expression datasets in FPKM and Counts formats with the clinic information of 487 PCa samples were collected from TCGA PCa dataset (https://portal.gdc.cancer.gov/, accessed on 1 June 2022) using the TCGA bio-links package, which was used for training set. Among these patients, 410 patients with complete BCR information were utilized for BCR-free survival. Another gene expression dataset (GSE70769) was mined from the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/, accessed on 1 June 2022) and used as the validation cohort. The overall bioinformatic analysis workflow in this study is presented in Figure 1.

2.2. Analysis and Screening of Differentially Expressed Genes (DEGs) between Low- and High-GS Groups

Among the 487 PCa patients, there were 291 cases with GS < 8 (low-GS group) and 196 cases with GS ≥ 8 (high-GS group). The patients’ clinical characteristics of the training dataset are shown in Supplementary Table S1. We analyzed the DEGs between low- and high-GS groups with the R package DEseq2, and the cut-off was set to adjusted p < 0.05 and |log2(fold change)| ≥ 0.5. The volcano map was developed using ‘ggplot2′ R package. Considering the biological significance and the convenient for further detection, those genes with mean FPKM less than 10 in tumor tissues were excluded from GS-associated DEGs.

2.3. Gene Oncology (GO) Analysis

We obtained the GO terms enrichment using ‘clusterProfiler’ R package with function ‘enrichGO’. The top 10 GO terms enriched in biological processes (BP) were visualized by ‘dotplot’ function.

2.4. Construction and Validation of the GS-Associated Gene Signature

BCR-free-survival-related genes were screened using Cox regression analysis based on the previously identified DEGs. We performed univariate Cox analysis using ‘Survival’ package in R and selected genes with p < 0.05 for next multivariate Cox regression analysis. The risk score was calculated using following formula: β1* Exp1 + β2* Exp2 + …. + βi* Expi. The β values were acquired by multivariate Cox regression analysis, and Exp was the level of indicated genes’ expression. The optimal cut-off values that could classify the patients in each dataset into high-risk and low-risk groups were obtained using ‘survivalROC’ package in R. Kaplan–Meier curves were plotted using ‘survfit’ function in ‘survival’ package, and p values were calculated with log rank test. The ‘survivalROC’ package of R software was performed to investigate the time dependence of the risk model.

2.5. Independent Prognostic Analysis

In order to assess whether risk score could be an independent risk factor, we performed univariate and multivariate Cox regression model analyses in training and testing cohorts. Variables with p < 0.05 in the multivariate Cox regression analysis were identified as independent risk factors.

2.6. Immune Microenvironment Analysis

Two deconvolution algorithms, namely CIBERSORT [23] and ESTIMATE [24], were performed to analyze the tumor microenvironment contexture. The relationship between the risk score and immune microenvironment was then assessed.

2.7. Box Plotting

Box plots were prepared using ‘ggboxplot’ function in the ‘ggpubr’ package of the R program. ‘Stat_compare_means’ function in the ‘ggpubr’ package of R program was used to determine the significance.

2.8. Statistical Analysis

Student t test was used to compare the expression of genes and risk score between indicated two groups. The BCR-free survival between indicated two groups was compared by Kaplan–Meier analysis, and p values were calculated with log rank test. Univariate Cox analysis and multivariate Cox regression analysis were performed to explore independent predictors of BCR-free survival. The hazard ratio (HR) and 95% confidence interval (CI) were calculated to compare the associated with BCR-free survival. ROC curves were implemented to identify the predictive accuracy of the risk signatures. All analyses were processed using the R software 4.2.1(The R Foundation, Vienna, Austria) and SPSS Version 26.0 (IBM, Armonk, NY, USA). If not specified above, p value less than 0.05 was considered statistically significant.

3. Results

3.1. The Identification and GO Enrichment Analysis of DEGs between PCa Patients with Low and High GS

Initially, we mined the DEGs based on Gleason score in TCGA-PCa datasets with a total of 487 prostate cancer patients. Among these, 196 cases had high GS (≥8), and 291 cases had low GS (<8). Then, we used |log2FC| ≥ 0.5 and p value < 0.05 as the screening criteria. A total of 127 upregulated and 133 downregulated genes were identified (Figure 2A). These upregulated and downregulated genes were used for GO biological processes (BP) analysis. The top 10 GO BP terms are shown in Figure 2B.

3.2. Identification of BCR Free Survival-Related Genes and Establishing the Prognostic Prediction Signature

Given that a gene must be expressed to some extent to be considered biologically important, together with the convenient for further detection, we selected 116 genes with mean FPKM greater than 10 in tumor tissues for further univariable regression analysis. We found that 32 genes were significantly associated with BCR-free survival (Table 1). Then, multivariable Cox regression analysis was performed, and a prediction model containing four genes was constructed as follows: (0.06447 ∗ expression of SFRP4) + (−0.06196 ∗ expression of FEV) + (0.37184 ∗ expression of COL1A1) + (0.06892 ∗ expression of SULF1). The risk score of each patient in the training set was calculated by the risk formula. The BCR status, risk score, and expression of the four genes in each patient from the training dataset are shown in Figure 3A,B. We also evaluated the BCR-free survival using the Kaplan–Meier method and log-rank test, and the results (Figure 3C) showed that the BCR-free survival time in the high-GS group was significantly shorter than that in the low-GS group (p < 0.0001). Furthermore, a time-dependent ROC curve was performed to evaluate the sensitivity and specificity of the four-gene signature for PFS prediction. Notably, the area under the ROC curve (AUC) was 0.66 for 1-year survival, 0.73 for 3-year survival, and 0.74 for 5-year survival (Figure 3D). Univariate Cox regression analysis and multivariate analysis demonstrated that the risk score was an independent predictor of poor clinical outcome in the training cohort (Table 2).

3.3. The Expression of Four Genes in Low-GS and High-GS, as Well as BCR and BCR Free Patients

We next assessed the expression of SFRP4, FEV, COL1A1, and SULF1 in low-GS and high-GS patients. The box plot depicted the expression of four genes in high-GS and low-GS groups. As shown in Figure 4A–D, the expression levels of SFRP4, COL1A1, and SULF1 were significantly upregulated in the high-GS group compared with in the low-GS group, while FEV expression was significantly downregulated in the high-GS group. Of note, the same expression pattern was observed in BCR and BCR-free patients. Among these four genes, FEV has a negative coefficient, indicating that it is a protective factor. SFRP4, COL1A1, and SULF1 have positive coefficients, suggesting negative effects on the prognosis. Intriguingly, FEV was highly expressed in BCR-free patients compared with in BCR patients. In contrast, the levels of SFRP4, COL1A1, and SULF1 were increased in BCR patients (Figure 5A–D).

3.4. The Association between Risk Score and Clinical Variables

The correlation between the risk score and clinical features was evaluated. The results showed that pathological T stage (Figure 6A), pathological N stage (Figure 6B), clinical T stage (Figure 6C), age (Figure 6D), and GS (Figure 6E) were significantly correlated with the risk score, indicating that the strong association between risk score and clinical pathological features.

3.5. The Correlation between Risk Score and Immune Microenvironment

Firstly, the CIBERSORT algorithm was used to analyze the correlation of risk score with immune signature. The results showed that tumors with high-risk scores were significantly correlated with high fractions of B cell naive, macrophages M1, macrophages M2, T cells CD4 memory resting, and T cells regulatory (Tregs) (Figure 7A). In the group with low-risk scores, there are high fractions of mast cell resting, NK cells activated, and plasma cells (Figure 7A). The abundance of each immune cell subtypes in PCa samples for the low- and high-risk groups is shown in Figure 7B. Moreover, ESTIMATE algorithms were applied to calculate the microenvironment score and immune score. The heatmap of the stromal score and immune score between the high- and low-risk groups is shown in Figure 7D. The box plots indicate that the low-risk group had a lower immune score and stromal score than those in the high-risk group (Figure 7C).

3.6. Validation of the Four-Gene Signature Using Another Independent Dataset

To investigate the reliability of the GS-associated four-gene signature, another independent dataset was used for further validation. Among the validation set, patients’ clinical characteristics are shown in Supplementary Table S2. We analyzed BCR status, risk score, and the four gene expression levels of each case, and the results are shown in Figure 8A,B. Of specific note, a substantially effective performance for BCR-free survival prediction was observed. Kaplan–Meier analysis exhibited that the patients in the high-risk group had obviously shorter BCR-free survival time than those in the low-risk group (Figure 8C). The AUCs for 1-year, 3-year, and 5-year survival are shown in Figure 8D. These results suggest that the four-gene signature is valid and reliable across datasets and platforms. Additionally, univariate Cox regression analysis and multivariate analysis showed that the risk score was an independent factor for predicting poor clinical outcome in the validation cohort (Table 3).

4. Discussion

Currently, PCa recurrence is a popular topic across the world. A large proportion of advanced PCa patients will recur [25], leading to ultimate death. Therefore, it is strongly desirable to effectively discover that patients with PCa have a high risk of BCR following RP. The utility of Gleason score in assessing prognosis at diagnosis is limited by the variability in tumor tissue grading [26], and the substantial heterogeneity of prognosis in patients with GS < 8 tumors [27]. Meanwhile, previous studies have revealed that signatures such as cell cycle progression genes [28], SigMuc1NW [29], lncRNAs [30] and microRNAs [31] have great potential as prognostic biomarkers. However, several concerns limit their prognostic and predictive power, such as inadequate samples and the lack of effective validation. Hence, exploring novel prediction models will greatly improve BCR prediction performance.
Our study predicts the recurrence of PCa after RP based on the GS score. We first identified DEGs between a high-GS group and a low-GS group, and then a univariable Cox analysis was carried out for screening BCR-associated genes. Then we used a multiple Cox regression model to establish a four-gene signature with prediction value. Then the risk score of each patient was calculated according to the formula. We found that BCR-free survival time in the high-GS group was significantly shorter than the low-GS group in each cohort. Time-dependent ROC analyses showed that the risk score had predictive power in both the training and validation sets. Among the four genes, the expression level of the gene (FEV) with negative coefficient was increased in BCR-free patients. Moreover, SFRP4, COL1A1, and SULF1 had positive coefficients, and their expression levels were upregulated in patients with BCR.
Previous studies have implicated these four genes in PCa. SFRP4 is one of the members of the secreted curl-related protein family (SFRP1–5) and is an extracellular inhibitor of Wnt signaling, whose role in carcinogenesis has been determined [32]. For PCa patients, the risk of BCR after RP increased with increased protein level of SFRP4 [33]. Research found that SFRP4 is one of the genes associated with PCa invasiveness and recurrence [33,34]. FEV is part of the ETS transcription factor family. Liang et al. found that its expression was significantly negatively associated with the GS [35]. The high expression of COL1A1 in colon cancer is significantly related to serous membrane infiltration, lymphatic metastasis, and hematogenous metastasis [36]. Liu et al. confirmed that COL1A1 is important in predicting PCa prognosis and progression [37]. Sulfatases (SULFs) have been locally acting tumor suppressors in many carcinomas [38]. Sulfatases 1 (SULF) are closely related to 6-Oendosulfatases [39], which are secreted or remain peripherally associated with proteoglycans at the cell membrane [40]. Some studies show that SULF1 suppresses the Wnt3A-driven growth of bone metastatic PCa [41]. Further studies exploring the clinical and biological significances of these genes in PCa will bring fresh insights into the etiology of the disease.
Bone is the most common site for prostate cancer to metastasize, and the incidence of advanced disease is 65–80% [42]. The skeletal microenvironment facilitates metastasis to PCa due to the complicated interactions between the microenvironment and tumor cells. Aside from cell precursors, bone marrow includes a variety of recirculating mature immune cells, such as dendritic cells (DC), macrophages, several T and B lymphocyte subsets, myeloid-derived suppressor cells (MDSCs), and NK cells. Some of these leukocytes are involved in pathogen clearance as well as anti-tumor actions [43,44]. Using CIBERSORT, we found a significant infiltration promotion of several immune cell subsets, particularly CD4+ T cells and macrophages M2, in high-risk groups. Intriguingly, this phenomenon increases the chance of metastasis and recurrence. In addition, we observed that the high-risk group has a significantly higher immune score and stromal score than the low-risk group, which showed that a distinct tumor microenvironment contexture exists between the two subtypes. Given the critical impact of the tumor immune microenvironment on metastasis, prompt and effective interventions to dysregulated immune cell infiltration can avert tumor recurrence.
There are several limitations in this study that need to be acknowledged. First, as this study was conducted using retrospective data that were obtained from public datasets, further prospective results are needed to support each other. Second, our study had a small number of cases with BCR or who died of PCa. Since BCR occurs over a wide time span, from a few months to over 15 years following the initial treatment [45], a long follow-up period is required to ascertain these outcome events. Third, future study is required to clarify the detailed molecular mechanism and function of these four genes in the development and progression of PCa.

5. Conclusions

Taken together, we conducted an integrated study to develop a GS-based four-gene signature (SFRP4, FEV, COL1A1, SULF1) for the prediction of the BCR of PCa patients after RP. The results from this study revealed a powerful prognostic indicator independent of and complementary to existing clinical factors for prognostication in PCa, such as the previously established Partin tables [46] and Kattan nomograms [47].

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm11237164/s1, Table S1. Clinical characteristics for training cohorts; Table S2. Clinical characteristics for validation cohorts.

Author Contributions

Conceptualization, Y.Y., J.C. and D.W.; methodology, Y.Y., X.Y. and H.G.; software, Y.Y. and H.G.; validation, Y.Y., J.C., D.W. and H.G.; formal analysis, H.G.; investigation, D.W., W.X. and C.L.; resources, Y.Y., H.G., D.W. and W.X.; data curation, Y.Y., X.Y. and H.G.; writing—original draft preparation, Y.Y., X.Y., D.W. and H.G.; writing—review and editing, Y.Y., H.G. and D.W.; visualization, J.C., C.L. and W.X.; supervision, Y.Y., H.G. and D.W.; project administration, H.G. and D.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Not applicable.

Acknowledgments

We thank all individuals for their participation in the study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jemal, A.; Siegel, R.; Ward, E.; Hao, Y.; Xu, J.; Thun, M.J. Cancer statistics, 2018. CA Cancer J. Clin. 2018, 68, 7–30. [Google Scholar]
  2. Venturini, N.J.; Drake, C.G. Immunotherapy for Prostate Cancer. Cold Spring Harb. Perspect. Med. 2019, 9. [Google Scholar] [CrossRef] [Green Version]
  3. Cao, W.; Chen, H.D.; Yu, Y.W.; Li, N.; Chen, W.Q. Changing profiles of cancer burden worldwide and in China: A secondary analysis of the global cancer statistics 2020. Chin. Med. J. 2021, 134, 783–791. [Google Scholar] [CrossRef] [PubMed]
  4. Chen, W.; Zheng, R.; Baade, P.D.; Zhang, S.; Zeng, H.; Bray, F.; Jemal, A.; Yu, X.Q.; He, J. Cancer statistics in China, 2015. CA Cancer J. Clin. 2016, 66, 115–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Yanagisawa, T.; Rajwa, P.; Thibault, C.; Gandaglia, G.; Mori, K.; Kawada, T.; Fukuokaya, W.; Shim, S.R.; Mostafaei, H.; Motlagh, R.S.; et al. Androgen Receptor Signaling Inhibitors in Addition to Docetaxel with Androgen Deprivation Therapy for Metastatic Hormone-sensitive Prostate Cancer: A Systematic Review and Meta-analysis. Eur. Urol. 2022, 82, 584–598. [Google Scholar] [CrossRef]
  6. Zhang, W.; Zhang, K. A transcriptomic signature for prostate cancer relapse prediction identified from the differentially expressed genes between TP53 mutant and wild-type tumors. Sci. Rep. 2022, 12, 10561. [Google Scholar] [CrossRef]
  7. Zhaoyong, F.; Bruce, J.T.; Elizabeth, B.H.; Michael, A.C.; Alan, W.P.; Patrick, C.W.; Mario, A.E. The natural history of metastatic progression in men with prostate-specific antigen recurrence after radical prostatectomy: Long-term follow-up. BJU Int. 2012, 109, 32–39. [Google Scholar]
  8. Moul, J.W. Prostate specific antigen only progression of prostate cancer. J. Urol. 2000, 163, 1632–1642. [Google Scholar] [CrossRef] [PubMed]
  9. Paller, C.J.; Antonarakis, E.S. Management of biochemically recurrent prostate cancer after local therapy: Evolving standards of care and new directions. Clin. Adv. Hematol. Oncol. 2013, 11, 14–23. [Google Scholar] [PubMed]
  10. Briganti, A.; Joniau, S.; Gandaglia, G.; Cozzarini, C.; Sun, M.; Tombal, B.; Wiegel, T. Patterns and predictors of early biochemical recurrence after radical prostatectomy and adjuvant radiation therapy in men with pT3N0 prostate cancer: Implications for multimodal therapies. Int. J. Radiat. Oncol. Biol. Phys. 2013, 87, 960–967. [Google Scholar] [CrossRef] [PubMed]
  11. Jinan, G.; Chenhui, Z.; Xinzhou, Z.; Zhong, W.; Tingting, C.; Jiashun, M.; Jinping, C.; Wenchuan, X.; Hao, C.; Mengli, H.; et al. A novel 8-gene panel for prediction of early biochemical recurrence in patients with prostate cancer after radical prostatectomy. Am. J. Cancer Res. 2022, 12, 3318–3332. [Google Scholar]
  12. D’Amico, A.V.; Whittington, R.; Malkowicz, S.B.; Schultz, D.; Blank, K.; Broderick, G.A.; Tomaszewski, J.E.; Renshaw, A.A.; Kaplan, I.; Beard, C.J.; et al. Biochemical outcome after radical prostatectomy, external beam radiation therapy, or interstitial radiation therapy for clinically localized prostate cancer. JAMA 1998, 280, 969–974. [Google Scholar] [CrossRef]
  13. Cooperberg, M.R.; Pasta, D.J.; Elkin, E.P.; Litwin, M.S.; Latini, D.M.; Du Chane, J.; Carroll, P.R. The University of California, San Francisco Cancer of the Prostate Risk Assessment score: A straightforward and reliable preoperative predictor of disease recurrence after radical prostatectomy. J. Urol. 2005, 173, 1938–1942. [Google Scholar] [CrossRef] [PubMed]
  14. Talantov, D.; Jatkoe, T.A.; Boehm, M.; Zhang, Y.; Ferguson, A.M.; Stricker, P.D.; Henshall, S.M. Gene based prediction of clinically localized prostate cancer progression after radical prostatectomy. J. Urol. 2010, 184, 1521–1528. [Google Scholar] [CrossRef] [PubMed]
  15. Yang, L.; Roberts, D.; Takhar, M.; Erho, N.; Bibby, B.A.; Thiruthaneeswaran, N.; West, C.M. Development and Validation of a 28-gene Hypoxia-related Prognostic Signature for Localized Prostate Cancer. EBioMedicine 2018, 31, 182–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Yuan, P.; Ling, L.; Fan, Q.; Gao, X.; Sun, T.; Miao, J.; Liu, B. A four-gene signature associated with clinical features can better predict prognosis in prostate cancer. Cancer Med. 2020, 9, 8202–8215. [Google Scholar] [CrossRef] [PubMed]
  17. Newhook, T.E.; Blais, E.M.; Lindberg, J.M.; Adair, S.J.; Xin, W.; Lee, J.K.; Bauer, T.W. A thirteen-gene expression signature predicts survival of patients with pancreatic cancer and identifies new genes of interest. PLoS ONE 2014, 9, e105631. [Google Scholar] [CrossRef] [PubMed]
  18. Knezevic, D.; Goddard, A.D.; Natraj, N.; Cherbavaz, D.B.; Clark-Langone, K.M.; Snable, J.; Watson, D.; Falzarano, S.M.; Magi-Galluzzi, C.; Klein, E.A.; et al. Analytical validation of the Oncotype DX prostate cancer assay—A clinical RT-PCR assay optimized for prostate needle biopsies. BMC Genom. 2013, 14, 690. [Google Scholar] [CrossRef] [Green Version]
  19. Chin, L.; Andersen, J.; Futreal, P.A. Futreal, Cancer genomics: From discovery science to personalized medicine. Nat. Med. 2011, 17, 297–303. [Google Scholar] [CrossRef]
  20. Bibikova, M.; Chudin, E.; Arsanjani, A.; Zhou, L.; Garcia, E.W.; Modder, J.; Kostelec, M.; Barker, D.; Downs, T.; Fan, J.-B.; et al. Expression signatures that correlated with Gleason score and relapse in prostate cancer. Genomics 2007, 89, 666–672. [Google Scholar] [CrossRef] [Green Version]
  21. Sinnott, J.A.; Peisch, S.F.; Tyekucheva, S.; Gerke, T.; Lis, R.; Rider, J.R.; Fiorentino, M.; Stampfer, M.J.; Mucci, L.A.; Loda, M.; et al. Prognostic Utility of a New mRNA Expression Signature of Gleason Score. Clin. Cancer Res. 2017, 23, 81–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Humphrey, P.A. Gleason grading and prognostic factors in carcinoma of the prostate. Mod. Pathol. 2004, 17, 292–306. [Google Scholar] [CrossRef] [Green Version]
  23. Newman, A.M.; Liu, C.L.; Green, M.R.; Gentles, A.J.; Feng, W.; Xu, Y.; Hoang, C.D.; Diehn, M.; Alizadeh, A.A. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods 2015, 12, 453–457. [Google Scholar] [CrossRef]
  24. Newman, A.M.; Liu, C.L.; Green, M.R.; Gentles, A.J.; Feng, W.; Xu, Y.; Hoang, C.D.; Diehn, M.; Alizadeh, A.A. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat. Commun. 2013, 4, 2612. [Google Scholar]
  25. Roth, S. Radiation with additional antiandrogen therapy in recurrent prostate cancer. Strahlenther. Onkol. 2017, 193, 679–681. [Google Scholar] [CrossRef] [PubMed]
  26. Allsbrook, W.C.; Mangold, K.; Johnson, M.H.; Lane, R.B.; Lane, C.G.; Amin, M.B.; Bostwick, D.G.; Humphrey, P.A.; Jones, E.C.; Reuter, V.E.; et al. Interobserver reproducibility of Gleason grading of prostatic carcinoma: Urologic pathologists. Hum. Pathol. 2001, 32, 74–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Sakr, W.A.; Tefilli, M.V.; Grignon, D.J.; Banerjee, M.; Dey, J.; Gheiler, E.L.; Tiguert, R.; Powell, I.J.; Wood, D.P. Gleason score 7 prostate cancer: A heterogeneous entity? Correlation with pathologic parameters and disease-free survival. Urology 2000, 56, 730–734. [Google Scholar] [CrossRef]
  28. Sommariva, S.; Tarricone, R.; Lazzeri, M.; Ricciardi, W.; Montorsi, F. Prognostic Value of the Cell Cycle Progression Score in Patients with Prostate Cancer: A Systematic Review and Meta-analysis. Eur. Urol. 2016, 69, 107–115. [Google Scholar] [CrossRef]
  29. Jiang, Y.; Mei, W.; Gu, Y.; Lin, X.; He, L.; Zeng, H.; Wei, F.; Wan, X.; Yang, H.; Major, P.; et al. Construction of a set of novel and robust gene expression signatures predicting prostate cancer recurrence. Mol. Oncol. 2018, 12, 1559–1578. [Google Scholar] [CrossRef] [Green Version]
  30. Shao, N.; Zhu, Y.; Wan, F.-N.; Ye, D.-W. Identification of seven long noncoding RNAs signature for prediction of biochemical recurrence in prostate cancer. Asian J. Androl. 2019, 21, 618–622. [Google Scholar]
  31. Zhao, Z.; Weickmann, S.; Jung, M.; Lein, M.; Kilic, E.; Stephan, C.; Jung, K. A Novel Predictor Tool of Biochemical Recurrence after Radical Prostatectomy Based on a Five-MicroRNA Tissue Signature. Cancers 2019, 11, 1603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Nusse, R.; Varmus, H.E. Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell 1982, 31, 99–109. [Google Scholar] [CrossRef] [PubMed]
  33. Mortensen, M.M.; Høyer, S.; Lynnerup, A.-S.; Ørntoft, T.F.; Sørensen, K.D.; Borre, M.; Dyrskjøt, L. Expression profiling of prostate cancer tissue delineates genes associated with recurrence after prostatectomy. Sci. Rep. 2015, 5, 16018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Klein, E.A.; Cooperberg, M.R.; Magi-Galluzzi, C.; Simko, J.P.; Falzarano, S.M.; Maddala, T.; Carroll, P.R. A 17-gene assay to predict prostate cancer aggressiveness in the context of gleason grade heterogeneity, tumor multifocality, and biopsy undersampling. Eur. Urol. 2014, 66, e117–e118. [Google Scholar] [CrossRef] [Green Version]
  35. Yu-Xiang, L.; Ying-Ke, L.; Zhi-Hao, Z.; Yang-Jia, Z.; Jian-Heng, Y.; Xue-Jin, Z.; Zhou-Da, C.; Zhuo-Yuan, L.; Ru-Jun, M.; Shu-Lin, W.; et al. Tumor Suppressor Role and Clinical Significance of the FEV Gene in Prostate Cancer. Dis. Markers 2022, 2022, 8724035. [Google Scholar]
  36. Zhang, Z.; Wang, Y.; Zhang, J.; Zhong, J.; Yang, R. COL1A1 promotes metastasis in colorectal cancer by regulating the WNT/PCP pathway. Mol. Med. Rep. 2018, 17, 5037–5042. [Google Scholar] [CrossRef]
  37. Liu, S.; He, B.; Li, H. Bisphenol S promotes the progression of prostate cancer by regulating the expression of COL1A1 and COL1A2. Toxicology 2022, 472, 153178. [Google Scholar] [CrossRef]
  38. Lai, J.-P.; Sandhu, D.S.; Shire, A.M.; Roberts, L.R. The tumor suppressor function of human sulfatase 1 (SULF1) in carcinogenesis. J. Gastrointest. Cancer 2008, 39, 149–158. [Google Scholar] [CrossRef] [Green Version]
  39. Ehammond, E.; Ekhurana, A.; Eshridhar, V.; Edredge, K. The Role of Heparanase and Sulfatases in the Modification of Heparan Sulfate Proteoglycans within the Tumor Microenvironment and Opportunities for Novel Cancer Therapeutics. Front. Oncol. 2014, 4, 195. [Google Scholar]
  40. Hossain, M.; Hosono-Fukao, T.; Tang, R.; Sugaya, N.; Van Kuppevelt, T.H.; Jenniskens, G.J.; Kimata, K.; Rosen, S.D.; Uchimura, K. Direct detection of HSulf-1 and HSulf-2 activities on extracellular heparan sulfate and their inhibition by PI-88. Glycobiology 2010, 20, 175–186. [Google Scholar] [CrossRef] [Green Version]
  41. Brasil da Costa, F.H.; Lewis, M.S.; Truong, A.; Carson, D.D.; Farach-Carson, M.C. SULF1 suppresses Wnt3A-driven growth of bone metastatic prostate cancer in perlecan-modified 3D cancer-stroma-macrophage triculture models. PLoS ONE 2020, 15, e0230354. [Google Scholar] [CrossRef] [PubMed]
  42. Tyekucheva, S.; Bowden, M.; Bango, C.; Giunchi, F.; Huang, Y.; Zhou, C.; Bondi, A.; Lis, R.; Van Hemelrijck, M.; Andrén, O.; et al. Stromal and epithelial transcriptional map of initiation progression and metastatic potential of human prostate cancer. Nat. Commun. 2017, 8, 420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Logothetis, C.; Morris, M.J.; Den, R.; Coleman, R.E. Current perspectives on bone metastases in castrate-resistant prostate cancer. Cancer Metastasis Rev. 2018, 37, 189–196. [Google Scholar] [CrossRef] [Green Version]
  44. Roato, I.; Vitale, M. The Uncovered Role of Immune Cells and NK Cells in the Regulation of Bone Metastasis. Front. Endocrinol. 2019, 10, 145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Marcin, P.; Jennifer, R.R.; Ove, A.; Sven-Olof, A.; Lars, H.; Hans-Olov, A.; Jan-Erik, J. Natural history of early, localized prostate cancer: A final report from three decades of follow-up. Eur. Urol. 2013, 63, 428–435. [Google Scholar]
  46. Eifler, J.B.; Feng, Z.; Lin, B.M.; Partin, M.T.; Humphreys, E.B.; Han, M.; Epstein, J.I.; Walsh, P.C.; Trock, B.J.; Partin, A.W. An updated prostate cancer staging nomogram (Partin tables) based on cases from 2006 to 2011. BJU Int. 2013, 111, 22–29. [Google Scholar] [CrossRef]
  47. Stephenson, A.J.; Kattan, M.W. Nomograms for prostate cancer. BJU Int. 2006, 98, 39–46. [Google Scholar] [CrossRef]
Figure 1. Flow chart of methods for building the Gleason Score (GS) based four-gene signature for prediction of BCR free survival in prostate cancer (PCa) patients; DEGs: differentially expressed genes.
Figure 1. Flow chart of methods for building the Gleason Score (GS) based four-gene signature for prediction of BCR free survival in prostate cancer (PCa) patients; DEGs: differentially expressed genes.
Jcm 11 07164 g001
Figure 2. Differentially expressed and functional enrichment analysis. (A) Volcano plot of DEGs between PCa patients with low and high GS. Red represents upregulated genes, and blue indicates downregulated genes. (B) Gene Ontology biological process analysis of the DEGs.
Figure 2. Differentially expressed and functional enrichment analysis. (A) Volcano plot of DEGs between PCa patients with low and high GS. Red represents upregulated genes, and blue indicates downregulated genes. (B) Gene Ontology biological process analysis of the DEGs.
Jcm 11 07164 g002
Figure 3. Construction of the four-gene signature in training cohort. (A,B) Distribution of risk score, BCR status, and gene expression of each case. (C) Kaplan–Meier curve analysis. (D) ROC curve analysis of the GS based four-gene signatures.
Figure 3. Construction of the four-gene signature in training cohort. (A,B) Distribution of risk score, BCR status, and gene expression of each case. (C) Kaplan–Meier curve analysis. (D) ROC curve analysis of the GS based four-gene signatures.
Jcm 11 07164 g003
Figure 4. The expression of four genes in high-GS and low-GS groups. The expression of SFRP4 (A), FEV (B), COL1A1 (C), and SULF1 (D) in PCa patients with high GS and low GS.
Figure 4. The expression of four genes in high-GS and low-GS groups. The expression of SFRP4 (A), FEV (B), COL1A1 (C), and SULF1 (D) in PCa patients with high GS and low GS.
Jcm 11 07164 g004
Figure 5. The expression of four genes in BCR and BCR-free groups. SFRP4 (A), FEV (B), COL1A1 (C), and SULF1 (D) expression levels in BCR and BCR-free PCa patients.
Figure 5. The expression of four genes in BCR and BCR-free groups. SFRP4 (A), FEV (B), COL1A1 (C), and SULF1 (D) expression levels in BCR and BCR-free PCa patients.
Jcm 11 07164 g005
Figure 6. The association between the GS-based four-gene signature and clinical characteristics of PCa patients. The distribution of risk scores was associated with pathologic T (A), clinical T (B), pathologic N (C), Gleason score (D), and age (E).
Figure 6. The association between the GS-based four-gene signature and clinical characteristics of PCa patients. The distribution of risk scores was associated with pathologic T (A), clinical T (B), pathologic N (C), Gleason score (D), and age (E).
Jcm 11 07164 g006
Figure 7. Immune infiltrating cell profile in PCa and correlation analysis. (A) The subgroup analysis of 22 immune infiltrating cells between the high-GS and low-GS groups. Red represents high-GS group while green represents low-GS group. (* p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001) (B) Bar plot of the proportion of immune infiltrating cells in the high- and low-risk PCa groups. (C) Box plots of immune score and stromal scores between the high- and low-risk groups. (D) Heat map of immune score and stromal scores between the high- and low-risk groups.
Figure 7. Immune infiltrating cell profile in PCa and correlation analysis. (A) The subgroup analysis of 22 immune infiltrating cells between the high-GS and low-GS groups. Red represents high-GS group while green represents low-GS group. (* p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001) (B) Bar plot of the proportion of immune infiltrating cells in the high- and low-risk PCa groups. (C) Box plots of immune score and stromal scores between the high- and low-risk groups. (D) Heat map of immune score and stromal scores between the high- and low-risk groups.
Jcm 11 07164 g007
Figure 8. Evaluating the prognostic power of the GS-based four-gene signature in the testing dataset (GSE70769). (A,B) Distribution of risk score, BCR status and gene expression of every patient in testing cohort. (C) ROC curve analysis of GS-based four-gene signature in testing cohort. (D) Kaplan–Meier curve analysis in testing cohort.
Figure 8. Evaluating the prognostic power of the GS-based four-gene signature in the testing dataset (GSE70769). (A,B) Distribution of risk score, BCR status and gene expression of every patient in testing cohort. (C) ROC curve analysis of GS-based four-gene signature in testing cohort. (D) Kaplan–Meier curve analysis in testing cohort.
Jcm 11 07164 g008
Table 1. Thirty-two genes significantly associated with the BCR free survival of patients in the training set (n = 487).
Table 1. Thirty-two genes significantly associated with the BCR free survival of patients in the training set (n = 487).
GeneHazard. Ratio (CI95)p Value
COL1A11.67 (1.37–2.03)0
BGN1.9 (1.46–2.46)1.00 × 10−6
SULF11.66 (1.34–2.06)5.00 × 10−6
COL3A11.65 (1.33–2.04)6.00 × 10−6
COL1A21.71 (1.35–2.16)7.00 × 10−6
COMP1.37 (1.18–1.59)2.90 × 10−5
POSTN1.55 (1.26–1.91)3.20 × 10−5
IGFBP31.71 (1.32–2.23)6.00 × 10−5
ELN1.7 (1.31–2.22)8.00 × 10−5
SFRP41.42 (1.19–1.71)0.000145
SFRP21.51 (1.22–1.87)0.000151
CDC42EP50.67 (0.54–0.83)0.000221
FN11.32 (1.13–1.53)0.000379
CXCL141.33 (1.13–1.56)0.000473
CAMK2N11.37 (1.14–1.63)0.000532
TK11.4 (1.14–1.71)0.001395
CKS21.42 (1.11–1.82)0.004743
CKM0.81 (0.7–0.94)0.005725
RHOU1.33 (1.08–1.63)0.006285
MSMB0.84 (0.74–0.95)0.006588
LBH1.28 (1.07–1.54)0.007336
ALDH1A11.2 (1.05–1.38)0.008648
FEV0.84 (0.73–0.96)0.010531
CRIP21.36 (1.07–1.72)0.01076
CHRNA20.84 (0.74–0.96)0.013084
PEBP40.85 (0.75–0.98)0.020598
VSIG20.87 (0.77–0.98)0.022015
KRT140.86 (0.75–0.98)0.029254
AZGP10.86 (0.75–0.99)0.033307
CPE0.75 (0.57–0.99)0.040177
LCN20.89 (0.79–1)0.043003
ACP30.84 (0.71–1)0.049737
Table 2. Univariate and multivariate analyses of the four-gene-based risk score and clinicopathological characteristics in TCGA-PCa training set.
Table 2. Univariate and multivariate analyses of the four-gene-based risk score and clinicopathological characteristics in TCGA-PCa training set.
UnivariableMultivariable
VariablesHR (95% CI)pHR (95% CI)p
Age1.303 (0.404, 4.203)0.658
Gleason Score0.032 (0.156, 0.584)<0.01
pT0.133 (0.041, 0.429)0.0010.218 (0.064, 0.747)0.015
pN0.199 (1.078, 3.705)0.028
Risk score0.270 (0.150, 0.487)<0.010.417 (0.222, 0.782)0.006
Table 3. Univariate and multivariate analyses of the four-gene-based risk score and clinicopathological characteristics in GEO70769 validation set.
Table 3. Univariate and multivariate analyses of the four-gene-based risk score and clinicopathological characteristics in GEO70769 validation set.
UnivariableMultivariable
VariablesHR (95% CI)pHR (95% CI)p
Gleason Score3.716 (1.829, 7.552)<0.012.773 (1.281, 6.005)0.01
pT3.915 (2.026, 7.562)<0.012.323 (1.115, 4.838)0.024
Surgical margins2.186 (1.185, 4.034)0.012
Prostate-specific antigen1.93 (1.04, 3.582)0.0372.254 (1.157, 4.392)0.017
Risk score0.331 (0.163, 0.675)0.0020.441 (0.206, 0.944)0.035
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yimamu, Y.; Yang, X.; Chen, J.; Luo, C.; Xiao, W.; Guan, H.; Wang, D. The Development of a Gleason Score-Related Gene Signature for Predicting the Prognosis of Prostate Cancer. J. Clin. Med. 2022, 11, 7164. https://doi.org/10.3390/jcm11237164

AMA Style

Yimamu Y, Yang X, Chen J, Luo C, Xiao W, Guan H, Wang D. The Development of a Gleason Score-Related Gene Signature for Predicting the Prognosis of Prostate Cancer. Journal of Clinical Medicine. 2022; 11(23):7164. https://doi.org/10.3390/jcm11237164

Chicago/Turabian Style

Yimamu, Yiliyasi, Xu Yang, Junxin Chen, Cheng Luo, Wenyang Xiao, Hongyu Guan, and Daohu Wang. 2022. "The Development of a Gleason Score-Related Gene Signature for Predicting the Prognosis of Prostate Cancer" Journal of Clinical Medicine 11, no. 23: 7164. https://doi.org/10.3390/jcm11237164

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop