Skip to main content
Log in

NMF-guided feature selection and genetic algorithm-driven framework for tumor mutational burden classification in bladder cancer using multi-omics data

  • Original Article
  • Published:
Network Modeling Analysis in Health Informatics and Bioinformatics Aims and scope Submit manuscript

Abstract

Accurately classifying bladder cancer patients based on Tumor Mutational Burden (TMB) is of paramount significance for prognosis and treatment decisions. To achieve that, we present a novel approach leveraging multi-omics data to differentiate between low and high TMB classes. The model combines feature selection and predictive modeling to unveil robust biomarkers associated with TMB classification. The Genetic Algorithm is employed to perform feature selection across DNA methylation, copy number alteration, and RNA-seq datasets. This process effectively reduces the dimensionality of the input data while retaining the most informative attributes. Subsequently, these selected features are projected into a latent space using non-negative matrix factorization, capturing the underlying patterns within the multi-omics data. Convolutional neural network among other machine learning machines to predict the class of TMB. The model introduces a promising classification power, showcasing the potential of these multi-omics biomarkers in accurately distinguishing between low and high TMB classes. The survival analysis reveals a substantial disparity between the cohorts classified as low-TMB and high-TMB. We propose a robust framework for TMB classification in bladder cancer that integrates multi-omics data, advanced machine learning techniques, and survival analysis to collectively pave the way for improved prognostic insights and personalized therapeutic interventions.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Data availability

The dataset analysed during the current study is available in the cBioPortal repository at https://www.cbioportal.org/study/summary?id=blca_tcga_pan_can_atlas_2018.

References

  • Alshomali L, Khorma R, Al-Refai A, Alkhateeb A (2023) Establishing a correlation of clinical characteristics with the level of tumor mutation burden in urothelial bladder carcinoma. In: 2023 IEEE international conference on bioinformatics and biomedicine (BIBM). IEEE, pp 3998–4001

  • Antoni S, Ferlay J, Soerjomataram I, Znaor A, Jemal A, Bray F (2016) Bladder cancer incidence and mortality: a global overview and recent trends. Eur Urol 71:96–108

    Article  Google Scholar 

  • Benjamini Y, Hochberg Y (1995) Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B (Methodol) 57:289–300

    Article  MathSciNet  Google Scholar 

  • Bladder cancer (2022) Mayo Clinic. https://www.mayoclinic.org/diseases-conditions/bladder-cancer/symptoms-causes/syc-20356104. Acessed 21 June 2023

  • Bladder cancer statistics: World cancer research fund international (2022) WCRF International. https://www.wcrf.org/cancer-trends/bladder-cancer-statistics/. Accessed 21 June 2023

  • Breiman L (2001) Random Forests. Mach Learn 45:5–32

    Article  Google Scholar 

  • Cerami E, Gao J, Dogrusoz U et al (2012) The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2:401–4

    Article  Google Scholar 

  • Chang K, Creighton CJ, Davis C et al (2013) The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 45:1113–20

    Article  Google Scholar 

  • Chen T, Guestrin C (2016) Xgboost: a scalable tree boosting system. In: Proceedings of the 22nd acm sigkdd international conference on knowledge discovery and data mining, pp 785–794

  • Dobruch J, Daneshmand S, Fisch M et al (2016) Gender and bladder cancer: a collaborative review of etiology, biology, and outcomes. Eur Urol 69:300–10

    Article  Google Scholar 

  • Elkarami B, Alkhateeb A, Rueda L (2016) Cost-sensitive classification on class-balanced ensembles for imbalanced non-coding RNA data. In: IEEE EMBS international student conference (ISC). IEEE, pp 1–4

  • Ferreira C (2001) Gene expression programming: a new adaptive algorithm for solving problems. arXiv preprint arXiv:cs/0102027

  • Fisher RA (1936) The use of multiple measurements in taxonomic problems. Ann Eugen 7:179–88

    Article  Google Scholar 

  • Gao J, Aksoy BA, Dogrusoz U et al (2013) Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6:l1

    Article  Google Scholar 

  • Ge SX, Jung D, Yao R (2019) ShinyGO: a graphical gene-set enrichment tool for animals and plants. Bioinformatics 36:2628–9

    Article  Google Scholar 

  • Goldberg DE (1989) Genetic algorithms in search, optimization, and machine learning. Addison-Wesley, Reading

    Google Scholar 

  • Hinton GE, Zemel R (1993) Autoencoders, minimum description length and Helmholtz free energy. Adv Neural Inf Process Syst 6

  • Holland JH (1992) Adaptation in natural and artificial systems: an introductory analysis with applications to biology, control, and artificial intelligence. MIT Press, Oxford

    Book  Google Scholar 

  • Kingma DP, Ba J (2014) Adam: a method for stochastic optimization. arXiv preprint arXiv:1412.6980

  • Lawlor RT, Mattiolo P, Mafficini A et al (2021) Tumor mutational burden as a potential biomarker for immunotherapy in pancreatic cancer: systematic review and still-open questions. Cancers (Basel) 13:3119

    Article  Google Scholar 

  • Lee DD, Seung HS (1999) Learning the parts of objects by non-negative matrix factorization. Nature 401:788–91

    Article  Google Scholar 

  • Lee D, Seung HS (2000) Algorithms for non-negative matrix factorization. In: Leen T, Dietterich T, Tresp V (eds) Advances in neural information processing systems. MIT Press. https://proceedings.neurips.cc/paper_files/paper/2000/file/f9d1152547c0bde01830b7e8bd60024c-Paper.pdf, p 13

  • Marcus L, Fashoyin-Aje LA, Donoghue M et al (2021) FDA approval summary: pembrolizumab for the treatment of tumor mutational burden-high solid tumors. Clin Cancer Res 27:4685–9

    Article  Google Scholar 

  • Min S, Lee B, Yoon S (2016) Deep learning in bioinformatics. Brief Bioinform 18:851–69

    Google Scholar 

  • Nair V, Hinton GE (2010) Rectified linear units improve restricted boltzmann machines. In: Proceedings of the 27th international conference on machine learning (ICML-10), pp 807–14

  • Pearson K (1901) On lines and planes of closest fit to systems of points in space. Philos Mag 2:559–72

    Article  Google Scholar 

  • Pedregosa F, Varoquaux G, Gramfort A et al (2011) Scikit-learn: machine learning in Python. J Mach Learn Res 12:2825–30

    MathSciNet  Google Scholar 

  • Ramalingam S, Hellmann M, Awad M et al (2018) Tumor mutational burden (TMB) as a biomarker for clinical benefit from dual immune checkpoint blockade with nivolumab (nivo)+ ipilimumab (ipi) in first-line (1L) non-small cell lung cancer (NSCLC): identification of TMB cutoff from CheckMate 568. Cancer Res 78:CT078–CT078

  • Sha D, Jin Z, Budczies J, Kluck K, Stenzinger A, Sinicrope FA (2020) Tumor mutational burden as a predictive biomarker in solid tumors. Cancer Discov 10:1808–25

    Article  Google Scholar 

  • Srivastava N, Hinton G, Krizhevsky A, Sutskever I, Salakhutdinov R (2014) Dropout: a simple way to prevent neural networks from overfitting. J Mach Learn Res 15:1929–58

    MathSciNet  Google Scholar 

  • Tang X, Qian WL, Yan WF, Pang T, Gong YL, Yang ZG (2021) Radiomic assessment as a method for predicting tumor mutation burden (TMB) of bladder cancer patients: a feasibility study. BMC Cancer 21:1–9

    Article  Google Scholar 

  • Thomas PD, Ebert D, Muruganujan A, Mushayahama T, Albou LP, Mi H (2022) PANTHER: making genome-scale phylogenetics accessible to all. Protein Sci 31:8–22

    Article  Google Scholar 

  • Yang J, Shi W, Yang Z et al (2023) Establishing a predictive model for tumor mutation burden status based on CT radiomics and clinical features of non-small cell lung cancer patients. Transl Lung Cancer Res 12:808–23

    Article  Google Scholar 

  • Zhang X, Wang J, Lu J et al (2021) Robust prognostic subtyping of muscle-invasive bladder cancer revealed by deep learning-based multi-omics data integration. Front Oncol 11:689626

    Article  Google Scholar 

Download references

Funding

This research was funded by the Scientific Research and Innovation Support Fund/Ministry of Higher Education and Scientific Research/Jordan, Grant number (ICT/1/16/2022). The recipients of this fund are Abedalrhman Alkhateeb and Hazem Qattous.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, AA, SA, and MA; Data curation, IA, and NA; Formal analysis, IA, LA, HQ, and AA; Funding acquisition, AA; Investigation, AA, LA, and SA; Methodology, IA, NA, AA, and MA; Project administration, AA, MA.

Corresponding author

Correspondence to Abedalrhman Alkhateeb.

Ethics declarations

Conflict of interest

The authors declare no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Al-Ghafer, I.A., AlAfeshat, N., Alshomali, L. et al. NMF-guided feature selection and genetic algorithm-driven framework for tumor mutational burden classification in bladder cancer using multi-omics data. Netw Model Anal Health Inform Bioinforma 13, 26 (2024). https://doi.org/10.1007/s13721-024-00460-7

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s13721-024-00460-7

Keywords

Navigation