C. M. Perou, T. Sorlie, and M. B. Eisen, Molecular portraits of human breast tumours, Nature, vol.406, pp.747-752, 2000.

F. Singh-dinesh, G. Phillip, and R. Kenneth, Gene expression correlates of clinical prostate cancer behavior Cancer Cell, vol.1, pp.203-209, 2002.

J. Van-'t-veer-laura, V. Dai-hongyue, and J. Marc, Gene expression profiling predicts clinical outcome of breast cancer Nature, vol.415, pp.530-536, 2002.

D. Anthony, V. , W. Richard, S. Malkowicz, and . Bruce, Biochemical outcome after radical prostatectomy, external beam radiation therapy, or interstitial radiation therapy for clinically localized prostate cancer Jama, vol.280, pp.969-974, 1998.

B. Marina, C. Eugene, and A. Amir, Expression signatures that correlated with Gleason score and relapse in prostate cancer Genomics, vol.89, pp.666-672, 2007.

L. Penney-kathryn, S. Jennifer, A. , and F. Katja, mRNA expression signature of Gleason grade predicts lethal prostate cancer, Journal of Clinical Oncology, vol.29, p.2391, 2011.

A. Sinnott-jennifer, P. Sam, F. , and T. Svitlana, Prognostic utility of a new mRNA expression signature of Gleason score Clinical Cancer Research, vol.23, pp.81-87, 2017.

J. Min, A. , G. Milan, S. , W. Jonathan et al., Gene expression signature of Gleason score is associated with prostate cancer outcomes in a radical prostatectomy cohort, Oncotarget, vol.8, p.43035, 2017.

B. Latil-alain, C. Ivan, and . Laurent, Gene Expression Profiling in Clinically Localized Prostate Cancer: A Four-Gene Expression Model Predicts Clinical Behavior Clinical Cancer Research, vol.9, pp.5477-5485, 2003.

Q. Long, J. Xu, and A. O. Osunkoya, Global Transcriptome Analysis of Formalin-Fixed Prostate Cancer Specimens Identifies Biomarkers of Disease Recurrence Cancer Research, vol.74, pp.3228-3237, 2014.

R. Shancheng, W. Gong-hong, and L. Dongbing, Whole-genome and transcriptome sequencing of prostate cancer identify new genetic alterations driving disease progression, European urology, vol.73, pp.322-339, 2018.

S. Ankit, H. Vincent, and L. Julie, The proteogenomic landscape of curable prostate cancer Cancer Cell, vol.35, pp.414-427, 2019.

E. Nicholas, C. Anamaria, and A. Vergara-ismael, Discovery and validation of a prostate cancer genomic classifier that predicts early metastasis following radical prostatectomy PloS one, vol.8, p.66855, 2013.

R. Karnes, B. Jeffrey, J. Eric, and . Davicioni-elai, Validation of a Genomic Classifier that Predicts Metastasis Following Radical Prostatectomy in an At Risk Patient Population, Journal of Urology, vol.190, pp.2047-2053, 2013.

A. Klein-eric, Y. Kasra, and H. Zaid, A genomic classifier improves prediction of metastatic disease within 5 years after surgery in node-negative high-risk prostate cancer patients managed by radical prostatectomy without adjuvant therapy, European Urology, vol.67, pp.778-786, 2015.

S. Ahva, J. Lewinger, R. Pablo, and . Jie, Novel Gene Expression Signature Predictive of Clinical Recurrence After Radical Prostatectomy in Early Stage Prostate Cancer Patients The Prostate, vol.76, pp.1239-1256, 2016.

M. Antonin and G. Daniel, Bridging the gap between reference and real transcriptomes Genome biology, vol.20, pp.1-7, 2019.

A. Jérôme, P. Nicolas, and C. Rayan, DE-kupl: exhaustive capture of biological variation in RNA-seq data through k-mer decomposition, Genome Biology, vol.18, p.243, 2017.

P. Marina, S. Zohra, and G. Mélina, Reference-free transcriptome exploration reveals novel RNAs for prostate cancer diagnosis Life Science Alliance, vol.2, pp.1-12, 2019.

T. Aubin, B. Sylvain, and B. Lucile, GECKO is a genetic algorithm to classify and explore high throughput sequencing data, Communications Biology, vol.2, p.222, 2019.

M. Stefan, K. Serge, and H. Catherine, Prediction of cancer outcome with microarrays: a multiple random validation strategy The Lancet, vol.365, pp.488-492, 2005.

L. Ein-dor, D. Zuk-or, and . Eytan, Thousands of samples are needed to generate a robust gene list for predicting outcome in cancer, Proceedings of the National Academy of Sciences, vol.103, pp.5923-5928, 2006.

S. Michiels, S. Koscielny, and C. Hill, Interpretation of microarray data in cancer, British Journal of Cancer, vol.96, pp.1155-1158, 2007.

V. David, D. Jacques, E. , and D. Vincent, Most random gene expression signatures are significantly associated with breast cancer outcome, PLoS computational biology, vol.7, p.1002240, 2011.

A. Adam, A. Jaeil, and A. Rehan, The molecular taxonomy of primary prostate cancer Cell, vol.163, pp.1011-1025, 2015.

F. Michael, S. Veronica, Y. , Y. Takafumi, and N. , Genomic hallmarks of localized, non-indolent prostate cancer, Nature, vol.541, pp.359-364, 2017.

S. Suzan, N. Ekaterina, and K. Yongsoo, Integrative epigenetic taxonomy of primary prostate cancer, Nature communications, vol.9, pp.1-12, 2018.

L. Jianfang, L. Tara, and A. Hoadley-katherine, An integrated TCGA pan-cancer clinical data resource to drive high-quality survival outcome analytics Cell, vol.173, pp.400-416, 2018.

A. Christophe, M. Geoffrey, and J. , Selection bias in gene extraction on the basis of microarray gene-expression data, Proceedings of the National Academy of Sciences, vol.99, pp.6562-6566, 2002.

M. Guillaume and K. Carl, A fast, lock-free approach for efficient parallel counting of occurrences of k-mers, Bioinformatics, vol.27, pp.764-770, 2011.

L. Bray-nicolas and P. Harold, Melsted Páll, Pachter Lior. Near-optimal probabilistic RNA-seq quantification Nature biotechnology, vol.34, pp.525-527, 2016.

R. Curtin-ryan, L. Marcus, and . Mikhail, Mentekidis Yannis, Ghaisas Sumedh, Zhang Shangtong. mlpack 3: a fast, flexible machine learning library, Journal of Open Source Software, vol.3, p.726, 2018.

F. Jerome, H. Trevor, and T. Robert, Regularization Paths for Generalized Linear Models via, Coordinate Descent Journal of Statistical Software, vol.33, pp.1-22, 2010.

M. Nicolai and B. Peter, Stability selection, Journal of the Royal Statistical Society: Series B (Statistical Methodology), vol.72, pp.417-473, 2010.

K. Max, Building Predictive Models in R Using the caret, Package Journal of Statistical Software, Articles, vol.28, pp.1-26, 2008.

R. J. Ensdb, Hsapiens.v79: Ensembl based annotation package 2017

. Liu-xian, R. Grogan-tristan, and H. Haley, Low CD38 identifies progenitor-like inflammation-associated luminal cells that can initiate human prostate cancer and predict poor outcome, Cell reports, vol.17, pp.2596-2606, 2016.

W. Tao, L. Zhuo, and G. Shuiming, The tumor suppressive role of CAMK2N1 in castration-resistant prostate cancer, Oncotarget, vol.5, p.3611, 2014.

L. Jing, S. Jia-xin, and W. Hua-tao, Collagen 1A1 (COL1A1) promotes metastasis of breast cancer and is a potential therapeutic target Discovery medicine, vol.25, pp.211-223, 2018.

W. Wu-xiaojuan, L. Hongbo, and . Yifan, GTSE1 promotes cell migration and invasion by regulating EMT in hepatocellular carcinoma and is associated with poor prognosis Scientific reports, vol.7, pp.1-12, 2017.

C. Chun-liang, M. Devalingam, and O. Pawel, Single-cell analysis of circulating tumor cells identifies cumulative expression patterns of EMT-related genes in metastatic prostate cancer The Prostate, vol.73, pp.813-826, 2013.

R. James, T. , T. Helga, and W. Wendy, Integrative genomics viewer, Nature biotechnology, vol.29, pp.24-30, 2011.

X. Wei, X. He, and L. Jia, Identification of low-density lipoprotein receptor class A domain containing 4 (LDLRAD4) as a prognostic indicator in primary gastrointestinal stromal tumors Current Problems in Cancer, p.100593, 2020.

M. Shaobo, Z. Long, and D. Weixing, Antisense lncRNA LDLRAD4-AS1 promotes metastasis by decreasing the expression of LDLRAD4 and predicts a poor prognosis in colorectal cancer Cell death & disease, vol.11, pp.1-16, 2020.

C. Charlie, D. , W. Derek, S. , and T. Chris, Molecular determinants of resistance to antiandrogen therapy Nature medicine, vol.10, pp.33-39, 2004.

C. Wei-yu, T. Yuan-chin, and Y. Hsiu-lien, Loss of SPDEF and gain of TGFBI activity after androgen deprivation therapy promote EMT and bone metastasis of prostate cancer, Science Signaling, vol.10, p.6826, 2017.

H. Leyten-gisele, H. Daphne, S. Frank, P. , J. Sander et al., Melchers Willem JG. Identification of a candidate gene panel for the early diagnosis of prostate cancer Clinical Cancer Research, vol.21, pp.3061-3070, 2015.

B. Marion, J. G. Bokhoven, A. , V. Gerald, and W. , DD3: a new prostatespecific gene, highly overexpressed in prostate cancer, Cancer research, vol.59, pp.5975-5984, 1999.

K. Georgios and S. Konstantinos, Avgeris Margaritis, et al. L-dopa decarboxylase (DDC) gene expression is related to outcome in patients with prostate cancer BJU international, vol.110, pp.267-273, 2012.

M. Ying, Z. , A. Joshua, and C. Goutam, A novel mechanism driving poor-prognosis prostate cancer: overexpression of the DNA repair gene, ribonucleotide reductase small subunit M2 (RRM2) Clinical Cancer Research, vol.25, pp.4480-4492, 2019.

Z. Wei-de, L. Yu-xiang, and L. Ying-ke, Tumor Suppressor Role and Clinical Implication of the Fifth Ewing Variant (FEV) Gene, an ETS Family Gene, Prostate Cancer Prostate Cancer, 2019.

M. Jennifer, M. Urszula, L. , L. Karen, and E. , The cancer-associated cell migration protein TSPAN1 is under control of androgens and its upregulation increases prostate cancer cell migration Scientific reports, vol.7, pp.1-11, 2017.

W. Song, X. Ming, and Z. Hai-bo, HMGCS2 functions as a tumor suppressor and has a prognostic impact in prostate cancer Pathology-Research and Practice, vol.215, p.152464, 2019.

A. Klein-eric, C. Matthew, R. Magi-galluzzi, and . Cristina, A 17-gene assay to predict prostate cancer aggressiveness in the context of gleason grade heterogeneity, tumor multifocality, and biopsy undersampling, European Urology, vol.66, pp.550-560, 2014.

W. Daniela, M. , and T. Robert, Survival analysis with high-dimensional covariates Statistical Methods in Medical Research, vol.19, pp.29-51, 2010.

R. Jorma, J. , R. Guillem, R. Sven, R. Sjoerd et al., Identifying subgroup markers in heterogeneous populations, Nucleic Acids Research, vol.41, pp.200-200, 2013.

F. Campos-laborie, A. Risueño, and M. Ortiz-estévez, DECO: decompose heterogeneous population cohorts for patient stratification and discovery of sample biomarkers using omic data profiling, Bioinformatics, vol.35, pp.3651-3662, 2019.