Novel deep learning-based solution for identification of prognostic subgroups in liver cancer (Hepatocellular carcinoma)

Background Liver cancer (Hepatocellular carcinoma; HCC) prevalence is increasing and with poor clinical outcome expected it means greater understanding of HCC aetiology is urgently required. This study explored a deep learning solution to detect biologically important features that distinguish prognostic subgroups. A novel architecture of an Artificial Neural Network (ANN) trained with a customised objective function (L RSC ) was developed. The ANN should discover new data representations, to detect patient subgroups that are biologically homogenous (clustering loss) and similar in survival (survival loss) while removing noise from the data (reconstruction loss). The model was applied to TCGA-HCC multi-omics data and benchmarked against baseline models that only use a reconstruction objective function (BCE, MSE) for learning. With the baseline models, the new features are then filtered based on survival information and used for clustering patients. Different variants of the customised objective function, incorporating only reconstruction and clustering losses (L RC ); and reconstruction and survival losses (L RS ) were also evaluated. Robust features consistently detected were compared between models and validated in TCGA and LIRI-JP HCC cohorts. Results The combined loss (L RSC ) discovered highly significant prognostic subgroups ( P -value = 1.55E−77) with more accurate sample assignment (Silhouette scores: 0.59–0.7) compared to baseline models (0.18–0.3). All L RSC bottleneck features (N = 100) were significant for survival, compared to only 11–21 for baseline models. Prognostic subgroups were not explained by disease grade or risk factors. Instead L RSC identified robust features including 377 mRNAs, many of which were novel (61.27%) compared to those identified by the other losses. Some 75 mRNAs were prognostic in TCGA, while 29 were prognostic in LIRI-JP also. L RSC also identified 15 robust miRNAs including two novel (hsa-let-7g; hsa-mir-550a-1) and 328 methylation features with 71% being prognostic. Gene-enrichment and Functional Annotation Analysis identified seven pathways differentiating prognostic clusters. Conclusions Combining cluster and survival metrics with the reconstruction objective function facilitated superior prognostic subgroup identification. The hybrid model identified more homogeneous clusters that consequently were more biologically meaningful. The novel and prognostic robust features extracted provide additional information to improve our understanding of a complex disease to help reveal its aetiology. Moreover, the gene features identified may have clinical applications as therapeutic targets.

[1]  H. El‐Serag,et al.  Epidemiology of viral hepatitis and hepatocellular carcinoma. , 2012, Gastroenterology.

[2]  Tieliu Shi,et al.  Deep Learning-Based Multi-Omics Data Integration Reveals Two Prognostic Subtypes in High-Risk Neuroblastoma , 2018, Front. Genet..

[3]  Keith A. Boroevich,et al.  Whole-genome mutational landscape and characterization of noncoding and structural mutations in liver cancer , 2016, Nature Genetics.

[4]  Yongliang Yuan,et al.  Large-scale analysis reveals a novel risk score to predict overall survival in hepatocellular carcinoma , 2018, Cancer management and research.

[5]  H. Kung,et al.  Hsa‐let‐7g inhibits proliferation of hepatocellular carcinoma cells by downregulation of c‐Myc and upregulation of p16INK4A , 2011, International journal of cancer.

[6]  Kumardeep Chaudhary,et al.  Deep Learning–Based Multi-Omics Integration Robustly Predicts Survival in Liver Cancer , 2017, Clinical Cancer Research.

[7]  Brad T. Sherman,et al.  Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources , 2008, Nature Protocols.

[8]  Cameron Davidson-Pilon,et al.  lifelines: survival analysis in Python , 2019, J. Open Source Softw..

[9]  P. Rousseeuw Silhouettes: a graphical aid to the interpretation and validation of cluster analysis , 1987 .

[10]  Kumardeep Chaudhary,et al.  Deep Learning data integration for better risk stratification models of bladder cancer , 2018, AMIA Joint Summits on Translational Science proceedings. AMIA Joint Summits on Translational Science.

[11]  Bo Yang,et al.  Towards K-means-friendly Spaces: Simultaneous Deep Learning and Clustering , 2016, ICML.

[12]  Gaël Varoquaux,et al.  Scikit-learn: Machine Learning in Python , 2011, J. Mach. Learn. Res..

[13]  M. Dumont,et al.  European Association for the Study of the Liver , 1971 .

[14]  C. Greene,et al.  ADAGE-Based Integration of Publicly Available Pseudomonas aeruginosa Gene Expression Data with Denoising Autoencoders Illuminates Microbe-Host Interactions , 2016, mSystems.

[15]  Hua Guo,et al.  Rictor promotes cell migration and actin polymerization through regulating ABLIM1 phosphorylation in Hepatocellular Carcinoma , 2020, International journal of biological sciences.

[16]  Joel Nothman,et al.  Author Correction: SciPy 1.0: fundamental algorithms for scientific computing in Python , 2020, Nature Methods.

[17]  P. Schirmacher,et al.  EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. , 2018, Journal of hepatology.

[18]  Daniel Rueckert,et al.  Deep learning cardiac motion analysis for human survival prediction , 2018, Nature Machine Intelligence.

[19]  Joel Nothman,et al.  SciPy 1.0-Fundamental Algorithms for Scientific Computing in Python , 2019, ArXiv.

[20]  Yongchang Zheng,et al.  DNA methylation-driven genes for constructing diagnostic, prognostic, and recurrence models for hepatocellular carcinoma , 2019, Theranostics.

[21]  J. Rowe,et al.  Review of hepatocellular carcinoma: Epidemiology, etiology, and carcinogenesis , 2017, Journal of carcinogenesis.

[22]  Yuan Ji,et al.  TCGA-Assembler 2: Software Pipeline for Retrieval and Processing of TCGA/CPTAC Data , 2017, bioRxiv.