1
|
Li X, Peng L, Wang YP, Zhang W. Open challenges and opportunities in federated foundation models towards biomedical healthcare. BioData Min 2025; 18:2. [PMID: 39755653 DOI: 10.1186/s13040-024-00414-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/09/2024] [Indexed: 01/06/2025] Open
Abstract
This survey explores the transformative impact of foundation models (FMs) in artificial intelligence, focusing on their integration with federated learning (FL) in biomedical research. Foundation models such as ChatGPT, LLaMa, and CLIP, which are trained on vast datasets through methods including unsupervised pretraining, self-supervised learning, instructed fine-tuning, and reinforcement learning from human feedback, represent significant advancements in machine learning. These models, with their ability to generate coherent text and realistic images, are crucial for biomedical applications that require processing diverse data forms such as clinical reports, diagnostic images, and multimodal patient interactions. The incorporation of FL with these sophisticated models presents a promising strategy to harness their analytical power while safeguarding the privacy of sensitive medical data. This approach not only enhances the capabilities of FMs in medical diagnostics and personalized treatment but also addresses critical concerns about data privacy and security in healthcare. This survey reviews the current applications of FMs in federated settings, underscores the challenges, and identifies future research directions including scaling FMs, managing data diversity, and enhancing communication efficiency within FL frameworks. The objective is to encourage further research into the combined potential of FMs and FL, laying the groundwork for healthcare innovations.
Collapse
Affiliation(s)
- Xingyu Li
- Department of Computer Science, Tulane University, New Orleans, LA, USA
| | - Lu Peng
- Department of Computer Science, Tulane University, New Orleans, LA, USA.
| | - Yu-Ping Wang
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - Weihua Zhang
- School of Computer Science, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Ballard JL, Wang Z, Li W, Shen L, Long Q. Deep learning-based approaches for multi-omics data integration and analysis. BioData Min 2024; 17:38. [PMID: 39358793 PMCID: PMC11446004 DOI: 10.1186/s13040-024-00391-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND The rapid growth of deep learning, as well as the vast and ever-growing amount of available data, have provided ample opportunity for advances in fusion and analysis of complex and heterogeneous data types. Different data modalities provide complementary information that can be leveraged to gain a more complete understanding of each subject. In the biomedical domain, multi-omics data includes molecular (genomics, transcriptomics, proteomics, epigenomics, metabolomics, etc.) and imaging (radiomics, pathomics) modalities which, when combined, have the potential to improve performance on prediction, classification, clustering and other tasks. Deep learning encompasses a wide variety of methods, each of which have certain strengths and weaknesses for multi-omics integration. METHOD In this review, we categorize recent deep learning-based approaches by their basic architectures and discuss their unique capabilities in relation to one another. We also discuss some emerging themes advancing the field of multi-omics integration. RESULTS Deep learning-based multi-omics integration methods were categorized broadly into non-generative (feedforward neural networks, graph convolutional neural networks, and autoencoders) and generative (variational methods, generative adversarial models, and a generative pretrained model). Generative methods have the advantage of being able to impose constraints on the shared representations to enforce certain properties or incorporate prior knowledge. They can also be used to generate or impute missing modalities. Recent advances achieved by these methods include the ability to handle incomplete data as well as going beyond the traditional molecular omics data types to integrate other modalities such as imaging data. CONCLUSION We expect to see further growth in methods that can handle missingness, as this is a common challenge in working with complex and heterogeneous data. Additionally, methods that integrate more data types are expected to improve performance on downstream tasks by capturing a comprehensive view of each sample.
Collapse
Affiliation(s)
- Jenna L Ballard
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA, 19104, USA.
| | - Zexuan Wang
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, 209 S. 33rd Street, Philadelphia, PA, 19104, USA
| | - Wenrui Li
- Department of Statistics, University of Connecticut, 215 Glenbrook Road, Storrs, CT, 06269, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, 423 Guardian Drive, Philadelphia, PA, 19104, USA.
| | - Qi Long
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, 423 Guardian Drive, Philadelphia, PA, 19104, USA.
| |
Collapse
|
3
|
Mathur A, Arya N, Pasupa K, Saha S, Roy Dey S, Saha S. Breast cancer prognosis through the use of multi-modal classifiers: current state of the art and the way forward. Brief Funct Genomics 2024; 23:561-569. [PMID: 38688724 DOI: 10.1093/bfgp/elae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 03/01/2024] [Accepted: 04/09/2024] [Indexed: 05/02/2024] Open
Abstract
We present a survey of the current state-of-the-art in breast cancer detection and prognosis. We analyze the evolution of Artificial Intelligence-based approaches from using just uni-modal information to multi-modality for detection and how such paradigm shift facilitates the efficacy of detection, consistent with clinical observations. We conclude that interpretable AI-based predictions and ability to handle class imbalance should be considered priority.
Collapse
Affiliation(s)
- Archana Mathur
- Department of Information Science and Engineering, Nitte Meenakshi Institute of Technology, Yelahanka, 560064, Karnataka, India
| | - Nikhilanand Arya
- School of Computer Engineering, Kalinga Institute of Industrial Technology, Deemed to be University, Bhubaneshwar, 751024, Odisha, India
| | - Kitsuchart Pasupa
- School of Information Technology, King Mongkut's Institute of Technology Ladkrabang, 1 Soi Chalongkrung 1, 10520, Bangkok, Thailand
| | - Sriparna Saha
- Computer Science and Engineering, Indian Institute of Technology Patna, Bihta, 801106, Bihar, India
| | - Sudeepa Roy Dey
- Department of Computer Science and Engineering, PES University, Hosur Road, 560100, Karnataka, India
| | - Snehanshu Saha
- CSIS and APPCAIR, BITS Pilani K.K Birla Goa Campus, Goa, 403726, Goa, India
- Div of AI Research, HappyMonk AI, Bangalore, 560078, Karnataka, India
| |
Collapse
|
4
|
Martins D, Abbasi M, Egas C, Arrais JP. Enhancing schizophrenia phenotype prediction from genotype data through knowledge-driven deep neural network models. Genomics 2024; 116:110910. [PMID: 39111546 DOI: 10.1016/j.ygeno.2024.110910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 08/20/2024]
Abstract
This article explores deep learning model design, drawing inspiration from the omnigenic model and genetic heterogeneity concepts, to improve schizophrenia prediction using genotype data. It introduces an innovative three-step approach leveraging neural networks' capabilities to efficiently handle genetic interactions. A locally connected network initially routes input data from variants to their corresponding genes. The second step employs an Encoder-Decoder to capture relationships among identified genes. The final model integrates knowledge from the first two and incorporates a parallel component to consider the effects of additional genes. This expansion enhances prediction scores by considering a larger number of genes. Trained models achieved an average AUC of 0.83, surpassing other genotype-trained models and matching gene expression dataset-based approaches. Additionally, tests on held-out sets reported an average sensitivity of 0.72 and an accuracy of 0.76, aligning with schizophrenia heritability predictions. Moreover, the study addresses genetic heterogeneity challenges by considering diverse population subsets.
Collapse
Affiliation(s)
- Daniel Martins
- Centre for Informatics and Systems of the University of Coimbra, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal; Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Maryam Abbasi
- Centre for Informatics and Systems of the University of Coimbra, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal; Polytechnic Institute of Coimbra, Applied Research Institute, Coimbra, Portugal; Research Centre for Natural Resources Environment and Society (CERNAS), Polytechnic Institute of Coimbra, Coimbra, Portugal.
| | - Conceição Egas
- Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Biocant - Transfer Technology Association, Cantanhede, Portugal; CNC - CNC Center for Neuroscience and Cell Biology, Coimbra, Portugal
| | - Joel P Arrais
- Centre for Informatics and Systems of the University of Coimbra, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
5
|
Li M, Cai Y, Zhang M, Deng S, Wang L. NNBGWO-BRCA marker: Neural Network and binary grey wolf optimization based Breast cancer biomarker discovery framework using multi-omics dataset. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 254:108291. [PMID: 38909399 DOI: 10.1016/j.cmpb.2024.108291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 05/09/2024] [Accepted: 06/16/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND AND OBJECTIVE Breast cancer is a multifaceted condition characterized by diverse features and a substantial mortality rate, underscoring the imperative for timely detection and intervention. The utilization of multi-omics data has gained significant traction in recent years to identify biomarkers and classify subtypes in breast cancer. This kind of research idea from part to whole will also be an inevitable trend in future life science research. Deep learning can integrate and analyze multi-omics data to predict cancer subtypes, which can further drive targeted therapies. However, there are few articles leveraging the nature of deep learning for feature selection. Therefore, this paper proposes a Neural Network and Binary grey Wolf Optimization based BReast CAncer bioMarker (NNBGWO-BRCAMarker) discovery framework using multi-omics data to obtain a series of biomarkers for precise classification of breast cancer subtypes. METHODS NNBGWO-BRCAMarker consists of two phases: in the first phase, relevant genes are selected using the weights obtained from a trained feedforward neural network; in the second phase, the binary grey wolf optimization algorithm is leveraged to further screen the selected genes, resulting in a set of potential breast cancer biomarkers. RESULTS The SVM classifier with RBF kernel achieved a classification accuracy of 0.9242 ± 0.03 when trained using the 80 biomarkers identified by NNBGWO-BRCAMarker, as evidenced by the experimental results. We conducted a comprehensive gene set analysis, prognostic analysis, and druggability analysis, unveiling 25 druggable genes, 16 enriched pathways strongly linked to specific subtypes of breast cancer, and 8 genes linked to prognostic outcomes. CONCLUSIONS The proposed framework successfully identified 80 biomarkers from the multi-omics data, enabling accurate classification of breast cancer subtypes. This discovery may offer novel insights for clinicians to pursue in further studies.
Collapse
Affiliation(s)
- Min Li
- School of Information Engineering, Nanchang Institute of Technology, No. 289 Tianxiang Road, Nanchang Jiangxi, PR China.
| | - Yuheng Cai
- School of Information Engineering, Nanchang Institute of Technology, No. 289 Tianxiang Road, Nanchang Jiangxi, PR China
| | - Mingzhuang Zhang
- School of Information Engineering, Nanchang Institute of Technology, No. 289 Tianxiang Road, Nanchang Jiangxi, PR China
| | - Shaobo Deng
- School of Information Engineering, Nanchang Institute of Technology, No. 289 Tianxiang Road, Nanchang Jiangxi, PR China
| | - Lei Wang
- School of Information Engineering, Nanchang Institute of Technology, No. 289 Tianxiang Road, Nanchang Jiangxi, PR China
| |
Collapse
|
6
|
Gunashekar DD, Bielak L, Oerther B, Benndorf M, Nedelcu A, Hickey S, Zamboglou C, Grosu AL, Bock M. Comparison of data fusion strategies for automated prostate lesion detection using mpMRI correlated with whole mount histology. Radiat Oncol 2024; 19:96. [PMID: 39080735 PMCID: PMC11287985 DOI: 10.1186/s13014-024-02471-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/14/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND In this work, we compare input level, feature level and decision level data fusion techniques for automatic detection of clinically significant prostate lesions (csPCa). METHODS Multiple deep learning CNN architectures were developed using the Unet as the baseline. The CNNs use both multiparametric MRI images (T2W, ADC, and High b-value) and quantitative clinical data (prostate specific antigen (PSA), PSA density (PSAD), prostate gland volume & gross tumor volume (GTV)), and only mp-MRI images (n = 118), as input. In addition, co-registered ground truth data from whole mount histopathology images (n = 22) were used as a test set for evaluation. RESULTS The CNNs achieved for early/intermediate / late level fusion a precision of 0.41/0.51/0.61, recall value of 0.18/0.22/0.25, an average precision of 0.13 / 0.19 / 0.27, and F scores of 0.55/0.67/ 0.76. Dice Sorensen Coefficient (DSC) was used to evaluate the influence of combining mpMRI with parametric clinical data for the detection of csPCa. We compared the DSC between the predictions of CNN's trained with mpMRI and parametric clinical and the CNN's trained with only mpMRI images as input with the ground truth. We obtained a DSC of data 0.30/0.34/0.36 and 0.26/0.33/0.34 respectively. Additionally, we evaluated the influence of each mpMRI input channel for the task of csPCa detection and obtained a DSC of 0.14 / 0.25 / 0.28. CONCLUSION The results show that the decision level fusion network performs better for the task of prostate lesion detection. Combining mpMRI data with quantitative clinical data does not show significant differences between these networks (p = 0.26/0.62/0.85). The results show that CNNs trained with all mpMRI data outperform CNNs with less input channels which is consistent with current clinical protocols where the same input is used for PI-RADS lesion scoring. TRIAL REGISTRATION The trial was registered retrospectively at the German Register for Clinical Studies (DRKS) under proposal number Nr. 476/14 & 476/19.
Collapse
Affiliation(s)
- Deepa Darshini Gunashekar
- Division of Medical Physics, Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.
| | - Lars Bielak
- Division of Medical Physics, Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Benedict Oerther
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matthias Benndorf
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andrea Nedelcu
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Samantha Hickey
- Division of Medical Physics, Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Constantinos Zamboglou
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Oncology Center, European University Cyprus, Limassol, Cyprus
| | - Anca-Ligia Grosu
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Michael Bock
- Division of Medical Physics, Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| |
Collapse
|
7
|
Chakraborty S, Sharma G, Karmakar S, Banerjee S. Multi-OMICS approaches in cancer biology: New era in cancer therapy. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167120. [PMID: 38484941 DOI: 10.1016/j.bbadis.2024.167120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 04/01/2024]
Abstract
Innovative multi-omics frameworks integrate diverse datasets from the same patients to enhance our understanding of the molecular and clinical aspects of cancers. Advanced omics and multi-view clustering algorithms present unprecedented opportunities for classifying cancers into subtypes, refining survival predictions and treatment outcomes, and unravelling key pathophysiological processes across various molecular layers. However, with the increasing availability of cost-effective high-throughput technologies (HTT) that generate vast amounts of data, analyzing single layers often falls short of establishing causal relations. Integrating multi-omics data spanning genomes, epigenomes, transcriptomes, proteomes, metabolomes, and microbiomes offers unique prospects to comprehend the underlying biology of complex diseases like cancer. This discussion explores algorithmic frameworks designed to uncover cancer subtypes, disease mechanisms, and methods for identifying pivotal genomic alterations. It also underscores the significance of multi-omics in tumor classifications, diagnostics, and prognostications. Despite its unparalleled advantages, the integration of multi-omics data has been slow to find its way into everyday clinics. A major hurdle is the uneven maturity of different omics approaches and the widening gap between the generation of large datasets and the capacity to process this data. Initiatives promoting the standardization of sample processing and analytical pipelines, as well as multidisciplinary training for experts in data analysis and interpretation, are crucial for translating theoretical findings into practical applications.
Collapse
Affiliation(s)
- Sohini Chakraborty
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Gaurav Sharma
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Sricheta Karmakar
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Satarupa Banerjee
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
8
|
Fawaz A, Ferraresi A, Isidoro C. Systems Biology in Cancer Diagnosis Integrating Omics Technologies and Artificial Intelligence to Support Physician Decision Making. J Pers Med 2023; 13:1590. [PMID: 38003905 PMCID: PMC10672164 DOI: 10.3390/jpm13111590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer is the second major cause of disease-related death worldwide, and its accurate early diagnosis and therapeutic intervention are fundamental for saving the patient's life. Cancer, as a complex and heterogeneous disorder, results from the disruption and alteration of a wide variety of biological entities, including genes, proteins, mRNAs, miRNAs, and metabolites, that eventually emerge as clinical symptoms. Traditionally, diagnosis is based on clinical examination, blood tests for biomarkers, the histopathology of a biopsy, and imaging (MRI, CT, PET, and US). Additionally, omics biotechnologies help to further characterize the genome, metabolome, microbiome traits of the patient that could have an impact on the prognosis and patient's response to the therapy. The integration of all these data relies on gathering of several experts and may require considerable time, and, unfortunately, it is not without the risk of error in the interpretation and therefore in the decision. Systems biology algorithms exploit Artificial Intelligence (AI) combined with omics technologies to perform a rapid and accurate analysis and integration of patient's big data, and support the physician in making diagnosis and tailoring the most appropriate therapeutic intervention. However, AI is not free from possible diagnostic and prognostic errors in the interpretation of images or biochemical-clinical data. Here, we first describe the methods used by systems biology for combining AI with omics and then discuss the potential, challenges, limitations, and critical issues in using AI in cancer research.
Collapse
Affiliation(s)
| | | | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (A.F.); (A.F.)
| |
Collapse
|
9
|
Yassi M, Chatterjee A, Parry M. Application of deep learning in cancer epigenetics through DNA methylation analysis. Brief Bioinform 2023; 24:bbad411. [PMID: 37985455 PMCID: PMC10661960 DOI: 10.1093/bib/bbad411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/08/2023] [Accepted: 10/25/2023] [Indexed: 11/22/2023] Open
Abstract
DNA methylation is a fundamental epigenetic modification involved in various biological processes and diseases. Analysis of DNA methylation data at a genome-wide and high-throughput level can provide insights into diseases influenced by epigenetics, such as cancer. Recent technological advances have led to the development of high-throughput approaches, such as genome-scale profiling, that allow for computational analysis of epigenetics. Deep learning (DL) methods are essential in facilitating computational studies in epigenetics for DNA methylation analysis. In this systematic review, we assessed the various applications of DL applied to DNA methylation data or multi-omics data to discover cancer biomarkers, perform classification, imputation and survival analysis. The review first introduces state-of-the-art DL architectures and highlights their usefulness in addressing challenges related to cancer epigenetics. Finally, the review discusses potential limitations and future research directions in this field.
Collapse
Affiliation(s)
- Maryam Yassi
- Department of Mathematics and Statistics, University of Otago, Dunedin, New Zealand
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- Honorary Professor, UPES University, Dehradun, India
| | - Matthew Parry
- Department of Mathematics and Statistics, University of Otago, Dunedin, New Zealand
- Te Pūnaha Matatini Centre of Research Excellence, University of Auckland, Auckland, New Zealand
| |
Collapse
|
10
|
Alatrany AS, Khan W, Hussain AJ, Mustafina J, Al-Jumeily D. Transfer Learning for Classification of Alzheimer's Disease Based on Genome Wide Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:2700-2711. [PMID: 37018274 DOI: 10.1109/tcbb.2022.3233869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Alzheimer's disease (AD) is a type of brain disorder that is regarded as a degenerative disease because the corresponding symptoms aggravate with the time progression. Single nucleotide polymorphisms (SNPs) have been identified as relevant biomarkers for this condition. This study aims to identify SNPs biomarkers associated with the AD in order to perform a reliable classification of AD. In contrast to existing related works, we utilize deep transfer learning with varying experimental analysis for reliable classification of AD. For this purpose, the convolutional neural networks (CNN) are firstly trained over the genome-wide association studies (GWAS) dataset requested from the AD neuroimaging initiative. We then employ the deep transfer learning for further training of our CNN (as base model) over a different AD GWAS dataset, to extract the final set of features. The extracted features are then fed into Support Vector Machine for classification of AD. Detailed experiments are performed using multiple datasets and varying experimental configurations. The statistical outcomes indicate an accuracy of 89% which is a significant improvement when benchmarked with existing related works.
Collapse
|
11
|
Kalemati M, Darvishi S, Koohi S. CapsNet-MHC predicts peptide-MHC class I binding based on capsule neural networks. Commun Biol 2023; 6:492. [PMID: 37147498 PMCID: PMC10162658 DOI: 10.1038/s42003-023-04867-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 04/24/2023] [Indexed: 05/07/2023] Open
Abstract
The Major Histocompatibility Complex (MHC) binds to the derived peptides from pathogens to present them to killer T cells on the cell surface. Developing computational methods for accurate, fast, and explainable peptide-MHC binding prediction can facilitate immunotherapies and vaccine development. Various deep learning-based methods rely on separate feature extraction from the peptide and MHC sequences and ignore their pairwise binding information. This paper develops a capsule neural network-based method to efficiently capture the peptide-MHC complex features to predict the peptide-MHC class I binding. Various evaluations confirmed our method outperformance over the alternative methods, while it can provide accurate prediction over less available data. Moreover, for providing precise insights into the results, we explored the essential features that contributed to the prediction. Since the simulation results demonstrated consistency with the experimental studies, we concluded that our method can be utilized for the accurate, rapid, and interpretable peptide-MHC binding prediction to assist biological therapies.
Collapse
Affiliation(s)
- Mahmood Kalemati
- Department of Computer Engineering, Sharif University of Technology, Tehran, Iran
| | - Saeid Darvishi
- Department of Computer Engineering, Sharif University of Technology, Tehran, Iran
| | - Somayyeh Koohi
- Department of Computer Engineering, Sharif University of Technology, Tehran, Iran.
| |
Collapse
|
12
|
Mohammed MA, Abdulkareem KH, Dinar AM, Zapirain BG. Rise of Deep Learning Clinical Applications and Challenges in Omics Data: A Systematic Review. Diagnostics (Basel) 2023; 13:diagnostics13040664. [PMID: 36832152 PMCID: PMC9955380 DOI: 10.3390/diagnostics13040664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
This research aims to review and evaluate the most relevant scientific studies about deep learning (DL) models in the omics field. It also aims to realize the potential of DL techniques in omics data analysis fully by demonstrating this potential and identifying the key challenges that must be addressed. Numerous elements are essential for comprehending numerous studies by surveying the existing literature. For example, the clinical applications and datasets from the literature are essential elements. The published literature highlights the difficulties encountered by other researchers. In addition to looking for other studies, such as guidelines, comparative studies, and review papers, a systematic approach is used to search all relevant publications on omics and DL using different keyword variants. From 2018 to 2022, the search procedure was conducted on four Internet search engines: IEEE Xplore, Web of Science, ScienceDirect, and PubMed. These indexes were chosen because they offer enough coverage and linkages to numerous papers in the biological field. A total of 65 articles were added to the final list. The inclusion and exclusion criteria were specified. Of the 65 publications, 42 are clinical applications of DL in omics data. Furthermore, 16 out of 65 articles comprised the review publications based on single- and multi-omics data from the proposed taxonomy. Finally, only a small number of articles (7/65) were included in papers focusing on comparative analysis and guidelines. The use of DL in studying omics data presented several obstacles related to DL itself, preprocessing procedures, datasets, model validation, and testbed applications. Numerous relevant investigations were performed to address these issues. Unlike other review papers, our study distinctly reflects different observations on omics with DL model areas. We believe that the result of this study can be a useful guideline for practitioners who look for a comprehensive view of the role of DL in omics data analysis.
Collapse
Affiliation(s)
- Mazin Abed Mohammed
- College of Computer Science and Information Technology, University of Anbar, Anbar 31001, Iraq
- eVIDA Lab, University of Deusto, 48007 Bilbao, Spain
- Correspondence: (M.A.M.); (B.G.Z.)
| | - Karrar Hameed Abdulkareem
- College of Agriculture, Al-Muthanna University, Samawah 66001, Iraq
- College of Engineering, University of Warith Al-Anbiyaa, Karbala 56001, Iraq
| | - Ahmed M. Dinar
- Computer Engineering Department, University of Technology- Iraq, Baghdad 19006, Iraq
| | | |
Collapse
|
13
|
Yu X, Zhou S, Zou H, Wang Q, Liu C, Zang M, Liu T. Survey of deep learning techniques for disease prediction based on omics data. HUMAN GENE 2023; 35:201140. [DOI: 10.1016/j.humgen.2022.201140] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
14
|
Li W, Wang S, Xu J, Xiang J. Inferring Latent MicroRNA-Disease Associations on a Gene-Mediated Tripartite Heterogeneous Multiplexing Network. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:3190-3201. [PMID: 35041612 DOI: 10.1109/tcbb.2022.3143770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
MicroRNA (miRNA) is a class of non-coding single-stranded RNA molecules encoded by endogenous genes with a length of about 22 nucleotides. MiRNAs have been successfully identified as differentially expressed in various cancers. There is evidence that disorders of miRNAs are associated with a variety of complex diseases. Therefore, inferring potential miRNA-disease associations (MDAs) is very important for understanding the aetiology and pathogenesis of many diseases and is useful to disease diagnosis, prognosis and treatment. First, We creatively fused multiple similarity subnetworks from multi-sources for miRNAs, genes and diseases by multiplexing technology, respectively. Then, three multiplexed biological subnetworks are connected through the extended binary association to form a tripartite complete heterogeneous multiplexed network (Tri-HM). Finally, because the constructed Tri-HM network can retain subnetworks' original topology and biological functions and expands the binary association and dependence between the three biological entities, rich neighbourhood information is obtained iteratively from neighbours by a non-equilibrium random walk. Through cross-validation, our tri-HM-RWR model obtained an AUC value of 0.8657, and an AUPR value of 0.2139 in the global 5-fold cross-validation, which shows that our model can more fully speculate disease-related miRNAs.
Collapse
|
15
|
MHDMF: Prediction of miRNA-disease associations based on Deep Matrix Factorization with Multi-source Graph Convolutional Network. Comput Biol Med 2022; 149:106069. [PMID: 36115300 DOI: 10.1016/j.compbiomed.2022.106069] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/31/2022] [Accepted: 08/27/2022] [Indexed: 11/24/2022]
Abstract
A growing number of works have proved that microRNAs (miRNAs) are a crucial biomarker in diverse bioprocesses affecting various diseases. As a good complement to high-cost wet experiment-based methods, numerous computational prediction methods have sprung up. However, there are still challenges that exist in making effective use of high false-negative associations and multi-source information for finding the potential associations. In this work, we develop an end-to-end computational framework, called MHDMF, which integrates the multi-source information on a heterogeneous network to discover latent disease-miRNA associations. Since high false-negative exist in the miRNA-disease associations, MHDMF utilizes the multi-source Graph Convolutional Network (GCN) to correct the false-negative association by reformulating the miRNA-disease association score matrix. The score matrix reformulation is based on different similarity profiles and known associations between miRNAs, genes, and diseases. Then, MHDMF employs Deep Matrix Factorization (DMF) to predict the miRNA-disease associations based on reformulated miRNA-disease association score matrix. The experimental results show that the proposed framework outperforms highly related comparison methods by a large margin on tasks of miRNA-disease association prediction. Furthermore, case studies suggest that MHDMF could be a convenient and efficient tool and may supply a new way to think about miRNA-disease association prediction.
Collapse
|
16
|
Leng D, Zheng L, Wen Y, Zhang Y, Wu L, Wang J, Wang M, Zhang Z, He S, Bo X. A benchmark study of deep learning-based multi-omics data fusion methods for cancer. Genome Biol 2022; 23:171. [PMID: 35945544 PMCID: PMC9361561 DOI: 10.1186/s13059-022-02739-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A fused method using a combination of multi-omics data enables a comprehensive study of complex biological processes and highlights the interrelationship of relevant biomolecules and their functions. Driven by high-throughput sequencing technologies, several promising deep learning methods have been proposed for fusing multi-omics data generated from a large number of samples. RESULTS In this study, 16 representative deep learning methods are comprehensively evaluated on simulated, single-cell, and cancer multi-omics datasets. For each of the datasets, two tasks are designed: classification and clustering. The classification performance is evaluated by using three benchmarking metrics including accuracy, F1 macro, and F1 weighted. Meanwhile, the clustering performance is evaluated by using four benchmarking metrics including the Jaccard index (JI), C-index, silhouette score, and Davies Bouldin score. For the cancer multi-omics datasets, the methods' strength in capturing the association of multi-omics dimensionality reduction results with survival and clinical annotations is further evaluated. The benchmarking results indicate that moGAT achieves the best classification performance. Meanwhile, efmmdVAE, efVAE, and lfmmdVAE show the most promising performance across all complementary contexts in clustering tasks. CONCLUSIONS Our benchmarking results not only provide a reference for biomedical researchers to choose appropriate deep learning-based multi-omics data fusion methods, but also suggest the future directions for the development of more effective multi-omics data fusion methods. The deep learning frameworks are available at https://github.com/zhenglinyi/DL-mo .
Collapse
Affiliation(s)
- Dongjin Leng
- Institute of Health Service and Transfusion Medicine, Beijing, People’s Republic of China
| | - Linyi Zheng
- School of Informatics, Xiamen University, Xiamen, People’s Republic of China
| | - Yuqi Wen
- Institute of Health Service and Transfusion Medicine, Beijing, People’s Republic of China
| | - Yunhao Zhang
- School of Informatics, Xiamen University, Xiamen, People’s Republic of China
| | - Lianlian Wu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, People’s Republic of China
| | - Jing Wang
- School of Medicine, Tsinghua University, Beijing, People’s Republic of China
| | - Meihong Wang
- School of Informatics, Xiamen University, Xiamen, People’s Republic of China
| | - Zhongnan Zhang
- School of Informatics, Xiamen University, Xiamen, People’s Republic of China
| | - Song He
- Institute of Health Service and Transfusion Medicine, Beijing, People’s Republic of China
| | - Xiaochen Bo
- Institute of Health Service and Transfusion Medicine, Beijing, People’s Republic of China
| |
Collapse
|
17
|
Wang HY, Zhao JP, Zheng CH, Su YS. scCNC: A method based on Capsule Network for Clustering scRNA-seq Data. Bioinformatics 2022; 38:3703-3709. [PMID: 35699473 DOI: 10.1093/bioinformatics/btac393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 05/28/2022] [Accepted: 06/11/2022] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION A large number of studies have shown that clustering is a crucial step in scRNA-seq analysis. Most existing methods are based on unsupervised learning without the prior exploitation of any domain knowledge, which does not utilize available gold-standard labels. When confronted by the high dimensionality and general dropout events of scRNA-seq data, purely unsupervised clustering methods may not produce biologically interpretable clusters, which complicates cell type assignment. RESULTS In this paper, we propose a semi-supervised clustering method based on a capsule network named scCNC, that integrates domain knowledge into the clustering step. Significantly, we also propose a Semi-supervised Greedy Iterative Training (SGIT) method used to train the whole network. Experiments on some real scRNA-seq datasets show that scCNC can significantly improve clustering performance and facilitate downstream analyses. AVAILABILITY The source code of scCNC is freely available at https://github.com/WHY-17/scCNC. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Hai-Yun Wang
- College of Mathematics and System Sciences, Xinjiang University, Urumqi, China
| | - Jian-Ping Zhao
- College of Mathematics and System Sciences, Xinjiang University, Urumqi, China.,Institute of Mathematics and Physics, Xinjiang University, Urumqi, China
| | - Chun-Hou Zheng
- College of Mathematics and System Sciences, Xinjiang University, Urumqi, China.,School of Artificial Intelligence, Anhui University, Hefei, China
| | - Yan-Sen Su
- School of Artificial Intelligence, Anhui University, Hefei, China
| |
Collapse
|
18
|
Mi JX, Feng J, Huang KY. Designing efficient convolutional neural network structure: A survey. Neurocomputing 2022. [DOI: 10.1016/j.neucom.2021.08.158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
19
|
|
20
|
Guo ZH, Chen ZH, You ZH, Wang YB, Yi HC, Wang MN. A learning-based method to predict LncRNA-disease associations by combining CNN and ELM. BMC Bioinformatics 2022; 22:622. [PMID: 35317723 PMCID: PMC8941737 DOI: 10.1186/s12859-022-04611-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/07/2021] [Indexed: 11/10/2022] Open
Abstract
Background lncRNAs play a critical role in numerous biological processes and life activities, especially diseases. Considering that traditional wet experiments for identifying uncovered lncRNA-disease associations is limited in terms of time consumption and labor cost. It is imperative to construct reliable and efficient computational models as addition for practice. Deep learning technologies have been proved to make impressive contributions in many areas, but the feasibility of it in bioinformatics has not been adequately verified. Results In this paper, a machine learning-based model called LDACE was proposed to predict potential lncRNA-disease associations by combining Extreme Learning Machine (ELM) and Convolutional Neural Network (CNN). Specifically, the representation vectors are constructed by integrating multiple types of biology information including functional similarity and semantic similarity. Then, CNN is applied to mine both local and global features. Finally, ELM is chosen to carry out the prediction task to detect the potential lncRNA-disease associations. The proposed method achieved remarkable Area Under Receiver Operating Characteristic Curve of 0.9086 in Leave-one-out cross-validation and 0.8994 in fivefold cross-validation, respectively. In addition, 2 kinds of case studies based on lung cancer and endometrial cancer indicate the robustness and efficiency of LDACE even in a real environment. Conclusions Substantial results demonstrated that the proposed model is expected to be an auxiliary tool to guide and assist biomedical research, and the close integration of deep learning and biology big data will provide life sciences with novel insights.
Collapse
Affiliation(s)
- Zhen-Hao Guo
- School of Electronics and Information Engineering, Tongji University, No. 4800 Cao'an Road, Shanghai, 201804, China
| | - Zhan-Heng Chen
- College of Computer Science and Engineering, Shenzhen University, Shenzhen, 518060, China.
| | - Zhu-Hong You
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710129, China
| | - Yan-Bin Wang
- College of Information Science and Engineering, Zaozhuang University, Zaozhuang, 277100, Shandong, China.
| | - Hai-Cheng Yi
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mei-Neng Wang
- School of Mathematics and Computer Science, Yichun University, Yichun, 336000, Jiangxi, China
| |
Collapse
|
21
|
Stahlschmidt SR, Ulfenborg B, Synnergren J. Multimodal deep learning for biomedical data fusion: a review. Brief Bioinform 2022; 23:bbab569. [PMID: 35089332 PMCID: PMC8921642 DOI: 10.1093/bib/bbab569] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/06/2021] [Accepted: 12/11/2021] [Indexed: 02/06/2023] Open
Abstract
Biomedical data are becoming increasingly multimodal and thereby capture the underlying complex relationships among biological processes. Deep learning (DL)-based data fusion strategies are a popular approach for modeling these nonlinear relationships. Therefore, we review the current state-of-the-art of such methods and propose a detailed taxonomy that facilitates more informed choices of fusion strategies for biomedical applications, as well as research on novel methods. By doing so, we find that deep fusion strategies often outperform unimodal and shallow approaches. Additionally, the proposed subcategories of fusion strategies show different advantages and drawbacks. The review of current methods has shown that, especially for intermediate fusion strategies, joint representation learning is the preferred approach as it effectively models the complex interactions of different levels of biological organization. Finally, we note that gradual fusion, based on prior biological knowledge or on search strategies, is a promising future research path. Similarly, utilizing transfer learning might overcome sample size limitations of multimodal data sets. As these data sets become increasingly available, multimodal DL approaches present the opportunity to train holistic models that can learn the complex regulatory dynamics behind health and disease.
Collapse
Affiliation(s)
| | | | - Jane Synnergren
- Systems Biology Research Center, University of Skövde, Sweden
| |
Collapse
|
22
|
Demirel HC, Arici MK, Tuncbag N. Computational approaches leveraging integrated connections of multi-omic data toward clinical applications. Mol Omics 2021; 18:7-18. [PMID: 34734935 DOI: 10.1039/d1mo00158b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In line with the advances in high-throughput technologies, multiple omic datasets have accumulated to study biological systems and diseases coherently. No single omics data type is capable of fully representing cellular activity. The complexity of the biological processes arises from the interactions between omic entities such as genes, proteins, and metabolites. Therefore, multi-omic data integration is crucial but challenging. The impact of the molecular alterations in multi-omic data is not local in the neighborhood of the altered gene or protein; rather, the impact diffuses in the network and changes the functionality of multiple signaling pathways and regulation of the gene expression. Additionally, multi-omic data is high-dimensional and has background noise. Several integrative approaches have been developed to accurately interpret the multi-omic datasets, including machine learning, network-based methods, and their combination. In this review, we overview the most recent integrative approaches and tools with a focus on network-based methods. We then discuss these approaches according to their specific applications, from disease-network and biomarker identification to patient stratification, drug discovery, and repurposing.
Collapse
Affiliation(s)
- Habibe Cansu Demirel
- Graduate School of Informatics, Middle East Technical University, Ankara, 06800, Turkey
| | - Muslum Kaan Arici
- Graduate School of Informatics, Middle East Technical University, Ankara, 06800, Turkey.,Foot and Mouth Diseases Institute, Ministry of Agriculture and Forestry, Ankara, 06044, Turkey
| | - Nurcan Tuncbag
- Chemical and Biological Engineering, College of Engineering, Koc University, Istanbul, 34450, Turkey.,School of Medicine, Koc University, Istanbul, 34450, Turkey.,Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.
| |
Collapse
|
23
|
Xiao Q, Dai J, Luo J. A survey of circular RNAs in complex diseases: databases, tools and computational methods. Brief Bioinform 2021; 23:6407737. [PMID: 34676391 DOI: 10.1093/bib/bbab444] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/21/2021] [Accepted: 09/28/2021] [Indexed: 01/22/2023] Open
Abstract
Circular RNAs (circRNAs) are a category of novelty discovered competing endogenous non-coding RNAs that have been proved to implicate many human complex diseases. A large number of circRNAs have been confirmed to be involved in cancer progression and are expected to become promising biomarkers for tumor diagnosis and targeted therapy. Deciphering the underlying relationships between circRNAs and diseases may provide new insights for us to understand the pathogenesis of complex diseases and further characterize the biological functions of circRNAs. As traditional experimental methods are usually time-consuming and laborious, computational models have made significant progress in systematically exploring potential circRNA-disease associations, which not only creates new opportunities for investigating pathogenic mechanisms at the level of circRNAs, but also helps to significantly improve the efficiency of clinical trials. In this review, we first summarize the functions and characteristics of circRNAs and introduce some representative circRNAs related to tumorigenesis. Then, we mainly investigate the available databases and tools dedicated to circRNA and disease studies. Next, we present a comprehensive review of computational methods for predicting circRNA-disease associations and classify them into five categories, including network propagating-based, path-based, matrix factorization-based, deep learning-based and other machine learning methods. Finally, we further discuss the challenges and future researches in this field.
Collapse
Affiliation(s)
- Qiu Xiao
- Hunan Normal University and Hunan Xiangjiang Artificial Intelligence Academy, Changsha, China
| | - Jianhua Dai
- Hunan Normal University and Hunan Xiangjiang Artificial Intelligence Academy, Changsha, China
| | - Jiawei Luo
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| |
Collapse
|
24
|
Tang B, Chen Y, Wang Y, Nie J. A Wavelet-Based Learning Model Enhances Molecular Prognosis in Pancreatic Adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2021; 2021:7865856. [PMID: 34697591 PMCID: PMC8541860 DOI: 10.1155/2021/7865856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/21/2021] [Indexed: 12/24/2022]
Abstract
Genome-wide omics technology boosts deep interrogation into the clinical prognosis and inherent mechanism of pancreatic oncology. Classic LASSO methods coequally treat all candidates, ignoring individual characteristics, thus frequently deteriorating performance with comparatively more predictors. Here, we propose a wavelet-based deep learning method in variable selection and prognosis formulation for PAAD with small samples and multisource information. With the genomic, epigenomic, and clinical cohort information from The Cancer Genome Atlas, the constructed five-molecule model is validated via Kaplan-Meier survival estimate, rendering significant prognosis capability on high- and low-risk subcohorts (p value < 0.0001), together with three predictors manifesting the individual prognosis significance (p value: 0.0012~0.024). Moreover, the performance of the prognosis model has been benchmarked against the traditional LASSO and wavelet-based methods in the 3- and 5-year prediction AUC items, respectively. Specifically, the proposed model with discrete stationary wavelet base (bior1.5) overwhelmingly outperformed traditional LASSO and wavelet-based methods (AUC: 0.787 vs. 0.782 and 0.721 for the 3-year case; AUC: 0.937 vs. 0.802 and 0.859 for the 5-year case). Thus, the proposed model provides a more accurate perspective, but with less predictor burden for clinical prognosis in the pancreatic carcinoma study.
Collapse
Affiliation(s)
- Binhua Tang
- Epigenetics & Function Group, Hohai University, Jiangsu 213022, China
| | - Yu Chen
- Epigenetics & Function Group, Hohai University, Jiangsu 213022, China
| | - Yuqi Wang
- Epigenetics & Function Group, Hohai University, Jiangsu 213022, China
| | - Jiafei Nie
- Epigenetics & Function Group, Hohai University, Jiangsu 213022, China
| |
Collapse
|
25
|
Vlachavas EI, Bohn J, Ückert F, Nürnberg S. A Detailed Catalogue of Multi-Omics Methodologies for Identification of Putative Biomarkers and Causal Molecular Networks in Translational Cancer Research. Int J Mol Sci 2021; 22:2822. [PMID: 33802234 PMCID: PMC8000236 DOI: 10.3390/ijms22062822] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
Recent advances in sequencing and biotechnological methodologies have led to the generation of large volumes of molecular data of different omics layers, such as genomics, transcriptomics, proteomics and metabolomics. Integration of these data with clinical information provides new opportunities to discover how perturbations in biological processes lead to disease. Using data-driven approaches for the integration and interpretation of multi-omics data could stably identify links between structural and functional information and propose causal molecular networks with potential impact on cancer pathophysiology. This knowledge can then be used to improve disease diagnosis, prognosis, prevention, and therapy. This review will summarize and categorize the most current computational methodologies and tools for integration of distinct molecular layers in the context of translational cancer research and personalized therapy. Additionally, the bioinformatics tools Multi-Omics Factor Analysis (MOFA) and netDX will be tested using omics data from public cancer resources, to assess their overall robustness, provide reproducible workflows for gaining biological knowledge from multi-omics data, and to comprehensively understand the significantly perturbed biological entities in distinct cancer types. We show that the performed supervised and unsupervised analyses result in meaningful and novel findings.
Collapse
Affiliation(s)
- Efstathios Iason Vlachavas
- Medical Informatics for Translational Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (J.B.); (F.Ü.)
| | - Jonas Bohn
- Medical Informatics for Translational Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (J.B.); (F.Ü.)
| | - Frank Ückert
- Medical Informatics for Translational Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (J.B.); (F.Ü.)
- Applied Medical Informatics, University Hospital Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Sylvia Nürnberg
- Medical Informatics for Translational Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (J.B.); (F.Ü.)
- Applied Medical Informatics, University Hospital Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
26
|
Biswas N, Chakrabarti S. Artificial Intelligence (AI)-Based Systems Biology Approaches in Multi-Omics Data Analysis of Cancer. Front Oncol 2020; 10:588221. [PMID: 33154949 PMCID: PMC7591760 DOI: 10.3389/fonc.2020.588221] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer is the manifestation of abnormalities of different physiological processes involving genes, DNAs, RNAs, proteins, and other biomolecules whose profiles are reflected in different omics data types. As these bio-entities are very much correlated, integrative analysis of different types of omics data, multi-omics data, is required to understanding the disease from the tumorigenesis to the disease progression. Artificial intelligence (AI), specifically machine learning algorithms, has the ability to make decisive interpretation of "big"-sized complex data and, hence, appears as the most effective tool for the analysis and understanding of multi-omics data for patient-specific observations. In this review, we have discussed about the recent outcomes of employing AI in multi-omics data analysis of different types of cancer. Based on the research trends and significance in patient treatment, we have primarily focused on the AI-based analysis for determining cancer subtypes, disease prognosis, and therapeutic targets. We have also discussed about AI analysis of some non-canonical types of omics data as they have the capability of playing the determiner role in cancer patient care. Additionally, we have briefly discussed about the data repositories because of their pivotal role in multi-omics data storing, processing, and analysis.
Collapse
Affiliation(s)
- Nupur Biswas
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, IICB TRUE Campus, Kolkata, India
| | - Saikat Chakrabarti
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, IICB TRUE Campus, Kolkata, India
| |
Collapse
|
27
|
Xi J, Yuan X, Wang M, Li A, Li X, Huang Q. Inferring subgroup-specific driver genes from heterogeneous cancer samples via subspace learning with subgroup indication. Bioinformatics 2020; 36:1855-1863. [PMID: 31626284 DOI: 10.1093/bioinformatics/btz793] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/23/2019] [Accepted: 10/16/2019] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION Detecting driver genes from gene mutation data is a fundamental task for tumorigenesis research. Due to the fact that cancer is a heterogeneous disease with various subgroups, subgroup-specific driver genes are the key factors in the development of precision medicine for heterogeneous cancer. However, the existing driver gene detection methods are not designed to identify subgroup specificities of their detected driver genes, and therefore cannot indicate which group of patients is associated with the detected driver genes, which is difficult to provide specifically clinical guidance for individual patients. RESULTS By incorporating the subspace learning framework, we propose a novel bioinformatics method called DriverSub, which can efficiently predict subgroup-specific driver genes in the situation where the subgroup annotations are not available. When evaluated by simulation datasets with known ground truth and compared with existing methods, DriverSub yields the best prediction of driver genes and the inference of their related subgroups. When we apply DriverSub on the mutation data of real heterogeneous cancers, we can observe that the predicted results of DriverSub are highly enriched for experimentally validated known driver genes. Moreover, the subgroups inferred by DriverSub are significantly associated with the annotated molecular subgroups, indicating its capability of predicting subgroup-specific driver genes. AVAILABILITY AND IMPLEMENTATION The source code is publicly available at https://github.com/JianingXi/DriverSub. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jianing Xi
- School of Mechanical Engineering , Northwestern Polytechnical University, Xi'an, 710072, China.,Center for OPTical IMagery Analysis and Learning (OPTIMAL), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Xiguo Yuan
- School of Computer Science and Technology, Xidian University, Xi'an 710071, China
| | - Minghui Wang
- School of Information Science and Technology, University of Science and Technology of China, Hefei 230027, China
| | - Ao Li
- School of Information Science and Technology, University of Science and Technology of China, Hefei 230027, China
| | - Xuelong Li
- Center for OPTical IMagery Analysis and Learning (OPTIMAL), Northwestern Polytechnical University, Xi'an, 710072, China.,School of Computer Science, Northwestern Polytechnical University, Xi'an 710072, China
| | - Qinghua Huang
- School of Mechanical Engineering , Northwestern Polytechnical University, Xi'an, 710072, China.,Center for OPTical IMagery Analysis and Learning (OPTIMAL), Northwestern Polytechnical University, Xi'an, 710072, China
| |
Collapse
|
28
|
Chierici M, Bussola N, Marcolini A, Francescatto M, Zandonà A, Trastulla L, Agostinelli C, Jurman G, Furlanello C. Integrative Network Fusion: A Multi-Omics Approach in Molecular Profiling. Front Oncol 2020; 10:1065. [PMID: 32714870 PMCID: PMC7340129 DOI: 10.3389/fonc.2020.01065] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
Recent technological advances and international efforts, such as The Cancer Genome Atlas (TCGA), have made available several pan-cancer datasets encompassing multiple omics layers with detailed clinical information in large collection of samples. The need has thus arisen for the development of computational methods aimed at improving cancer subtyping and biomarker identification from multi-modal data. Here we apply the Integrative Network Fusion (INF) pipeline, which combines multiple omics layers exploiting Similarity Network Fusion (SNF) within a machine learning predictive framework. INF includes a feature ranking scheme (rSNF) on SNF-integrated features, used by a classifier over juxtaposed multi-omics features (juXT). In particular, we show instances of INF implementing Random Forest (RF) and linear Support Vector Machine (LSVM) as the classifier, and two baseline RF and LSVM models are also trained on juXT. A compact RF model, called rSNFi, trained on the intersection of top-ranked biomarkers from the two approaches juXT and rSNF is finally derived. All the classifiers are run in a 10x5-fold cross-validation schema to warrant reproducibility, following the guidelines for an unbiased Data Analysis Plan by the US FDA-led initiatives MAQC/SEQC. INF is demonstrated on four classification tasks on three multi-modal TCGA oncogenomics datasets. Gene expression, protein expression and copy number variants are used to predict estrogen receptor status (BRCA-ER, N = 381) and breast invasive carcinoma subtypes (BRCA-subtypes, N = 305), while gene expression, miRNA expression and methylation data is used as predictor layers for acute myeloid leukemia and renal clear cell carcinoma survival (AML-OS, N = 157; KIRC-OS, N = 181). In test, INF achieved similar Matthews Correlation Coefficient (MCC) values and 97% to 83% smaller feature sizes (FS), compared with juXT for BRCA-ER (MCC: 0.83 vs. 0.80; FS: 56 vs. 1801) and BRCA-subtypes (0.84 vs. 0.80; 302 vs. 1801), improving KIRC-OS performance (0.38 vs. 0.31; 111 vs. 2319). INF predictions are generally more accurate in test than one-dimensional omics models, with smaller signatures too, where transcriptomics consistently play the leading role. Overall, the INF framework effectively integrates multiple data levels in oncogenomics classification tasks, improving over the performance of single layers alone and naive juxtaposition, and provides compact signature sizes.
Collapse
Affiliation(s)
| | - Nicole Bussola
- Fondazione Bruno Kessler, Trento, Italy
- University of Trento, Trento, Italy
| | | | - Margherita Francescatto
- Fondazione Bruno Kessler, Trento, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | | | | | | | | | | |
Collapse
|
29
|
Xiao Q, Zhang N, Luo J, Dai J, Tang X. Adaptive multi-source multi-view latent feature learning for inferring potential disease-associated miRNAs. Brief Bioinform 2020; 22:2043-2057. [PMID: 32186712 DOI: 10.1093/bib/bbaa028] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/16/2020] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
Accumulating evidence has shown that microRNAs (miRNAs) play crucial roles in different biological processes, and their mutations and dysregulations have been proved to contribute to tumorigenesis. In silico identification of disease-associated miRNAs is a cost-effective strategy to discover those most promising biomarkers for disease diagnosis and treatment. The increasing available omics data sources provide unprecedented opportunities to decipher the underlying relationships between miRNAs and diseases by computational models. However, most existing methods are biased towards a single representation of miRNAs or diseases and are also not capable of discovering unobserved associations for new miRNAs or diseases without association information. In this study, we present a novel computational method with adaptive multi-source multi-view latent feature learning (M2LFL) to infer potential disease-associated miRNAs. First, we adopt multiple data sources to obtain similarity profiles and capture different latent features according to the geometric characteristic of miRNA and disease spaces. Then, the multi-modal latent features are projected to a common subspace to discover unobserved miRNA-disease associations in both miRNA and disease views, and an adaptive joint graph regularization term is developed to preserve the intrinsic manifold structures of multiple similarity profiles. Meanwhile, the Lp,q-norms are imposed into the projection matrices to ensure the sparsity and improve interpretability. The experimental results confirm the superior performance of our proposed method in screening reliable candidate disease miRNAs, which suggests that M2LFL could be an efficient tool to discover diagnostic biomarkers for guiding laborious clinical trials.
Collapse
|
30
|
Wang Y, Huang L, Jiang S, Wang Y, Zou J, Fu H, Yang S. Capsule Networks Showed Excellent Performance in the Classification of hERG Blockers/Nonblockers. Front Pharmacol 2020; 10:1631. [PMID: 32063849 PMCID: PMC6997788 DOI: 10.3389/fphar.2019.01631] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/13/2019] [Indexed: 02/05/2023] Open
Abstract
Capsule networks (CapsNets), a new class of deep neural network architectures proposed recently by Hinton et al., have shown a great performance in many fields, particularly in image recognition and natural language processing. However, CapsNets have not yet been applied to drug discovery-related studies. As the first attempt, we in this investigation adopted CapsNets to develop classification models of hERG blockers/nonblockers; drugs with hERG blockade activity are thought to have a potential risk of cardiotoxicity. Two capsule network architectures were established: convolution-capsule network (Conv-CapsNet) and restricted Boltzmann machine-capsule networks (RBM-CapsNet), in which convolution and a restricted Boltzmann machine (RBM) were used as feature extractors, respectively. Two prediction models of hERG blockers/nonblockers were then developed by Conv-CapsNet and RBM-CapsNet with the Doddareddy's training set composed of 2,389 compounds. The established models showed excellent performance in an independent test set comprising 255 compounds, with prediction accuracies of 91.8 and 92.2% for Conv-CapsNet and RBM-CapsNet models, respectively. Various comparisons were also made between our models and those developed by other machine learning methods including deep belief network (DBN), convolutional neural network (CNN), multilayer perceptron (MLP), support vector machine (SVM), k-nearest neighbors (kNN), logistic regression (LR), and LightGBM, and with different training sets. All the results showed that the models by Conv-CapsNet and RBM-CapsNet are among the best classification models. Overall, the excellent performance of capsule networks achieved in this investigation highlights their potential in drug discovery-related studies.
Collapse
Affiliation(s)
- Yiwei Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- College of Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Lei Huang
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China
- Basic Teaching Department, Sichuan College of Architectural Technology, Deyang, China
| | - Siwen Jiang
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Yifei Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Zou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongguang Fu
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|