1
|
Hu G, Zhou T, Zhou P, Yau SST. Novel natural vector with asymmetric covariance for classifying biological sequences. Gene 2025:149532. [PMID: 40367998 DOI: 10.1016/j.gene.2025.149532] [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: 01/12/2025] [Revised: 04/07/2025] [Accepted: 04/23/2025] [Indexed: 05/16/2025]
Abstract
The genome sequences of organisms form a large and complex landscape, presenting a significant challenge in bioinformatics: how to utilize mathematical tools to describe and analyze this space effectively. The ability to compare relationships between different organisms depends on creating a rational mapping rule that can uniformly encode genome sequences of varying lengths as vectors in a measurable space. This mapping would enable researchers to apply modern mathematical and machine learning techniques to otherwise challenging genomic comparisons. The natural vector method has been proposed as a concise and effective approach to accomplish this. However, its various iterations have certain limitations. In response, we carefully analyze the strengths and weaknesses of these natural vector methods and propose an improved version-an asymmetric covariance natural vector method (ACNV). This new method incorporates k-mer information alongside covariance computations with asymmetric properties between base positions. We tested ACNV on microbial genome sequence datasets, including bacterial, fungal, and viral sequences, evaluating its performance in terms of classification accuracy and convex hull separation. The results demonstrate that ACNV effectively captures sequence characteristics, showcasing its robust sequence representation capabilities and highlighting its elegant geometric properties.
Collapse
Affiliation(s)
- Guoqing Hu
- Beijing Institute of Mathematical Sciences and Applications (BIMSA), 101408, Beijing, China.
| | - Tao Zhou
- Department of Mathematical Sciences, Tsinghua University, 100084, Beijing, China
| | - Piyu Zhou
- School of Mathematical Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Stephen Shing-Toung Yau
- Beijing Institute of Mathematical Sciences and Applications (BIMSA), 101408, Beijing, China.
| |
Collapse
|
2
|
Qi H, Zhao H, Li E, Lu X, Yu N, Liu J, Han J. DeepQA: A Unified Transcriptome-Based Aging Clock Using Deep Neural Networks. Aging Cell 2025; 24:e14471. [PMID: 39757434 PMCID: PMC12074024 DOI: 10.1111/acel.14471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/21/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025] Open
Abstract
Understanding the complex biological process of aging is of great value, especially as it can help develop therapeutics to prolong healthy life. Predicting biological age from gene expression data has shown to be an effective means to quantify aging of a subject, and to identify molecular and cellular biomarkers of aging. A typical approach for estimating biological age, adopted by almost all existing aging clocks, is to train machine learning models only on healthy subjects, but to infer on both healthy and unhealthy subjects. However, the inherent bias in this approach results in inaccurate biological age as shown in this study. Moreover, almost all existing transcriptome-based aging clocks were built around an inefficient procedure of gene selection followed by conventional machine learning models such as elastic nets, linear discriminant analysis etc. To address these limitations, we proposed DeepQA, a unified aging clock based on mixture of experts. Unlike existing methods, DeepQA is equipped with a specially designed Hinge-Mean-Absolute-Error (Hinge-MAE) loss so that it can train on both healthy and unhealthy subjects of multiple cohorts to reduce the bias of inferring biological age of unhealthy subjects. Our experiments showed that DeepQA significantly outperformed existing methods for biological age estimation on both healthy and unhealthy subjects. In addition, our method avoids the inefficient exhaustive search of genes, and provides a novel means to identify genes activated in aging prediction, alternative to such as differential gene expression analysis.
Collapse
Affiliation(s)
- Hongqian Qi
- State Key Laboratory of Medicinal Chemical BiologyNankai UniversityTianjinChina
- College of PharmacyNankai UniversityTianjinChina
| | - Hongchen Zhao
- College of Artificial IntelligenceNankai UniversityTianjinChina
| | - Enyi Li
- College of Artificial IntelligenceNankai UniversityTianjinChina
| | - Xinyi Lu
- State Key Laboratory of Medicinal Chemical BiologyNankai UniversityTianjinChina
| | - Ningbo Yu
- College of Artificial IntelligenceNankai UniversityTianjinChina
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of EducationNankai UniversityChina
| | - Jinchao Liu
- College of Artificial IntelligenceNankai UniversityTianjinChina
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of EducationNankai UniversityChina
| | - Jianda Han
- College of Artificial IntelligenceNankai UniversityTianjinChina
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of EducationNankai UniversityChina
| |
Collapse
|
3
|
Gao X, Ma S, Ni W, Kuang Y, Yu Y, Zhou L, Li Y, Guo C, Xu C, Li L, Huang H, Han J. Design of Multi-Cancer VOCs Profiling Platform via a Deep Learning-Assisted Sensing Library Screening Strategy. Anal Chem 2025; 97:8301-8312. [PMID: 40211116 DOI: 10.1021/acs.analchem.4c06468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
The efficiency of sensor arrays in parallel discrimination of multianalytes is fundamentally influenced by the quantity and performance of the sensor elements. The advent of combinational design has notably accelerated the generation of chemical libraries, offering numerous candidates for the development of robust sensor arrays. However, screening elements with superior cross-responsiveness remains challenging, impeding the development of high-performance sensor arrays. Herein, we propose a new deep learning-assisted, two-step screening strategy to identify the optimal combination of minimal sensor elements, using a designed volatile organic compounds (VOCs)-targeted sensor library. 400 sensing elements constructed by pairing 20 ionizable cationic elements and 20 anionic dyes in the sensor library were employed for various VOCs, generating plentiful color variation data. By employing a feedforward neural network─random forest-recursive feature elimination (FRR) algorithm, sensing elements were effectively screened, resulting in the rapidly producing 8-element and 10-element arrays for two VOC models, both achieving 100% discrimination accuracy. Furthermore, a smartphone-based point-of-care testing (POCT) platform achieved cancer discrimination in a simulated cancer VOC model, using image-based deep learning, demonstrating the rationality and practicality of deep learning in the assembly of sensor elements for parallel sensing platforms.
Collapse
Affiliation(s)
- Xu Gao
- State Key Laboratory of Natural Medicines, National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Shuoyang Ma
- State Key Laboratory of Natural Medicines, National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Weiwei Ni
- State Key Laboratory of Natural Medicines, National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yongbin Kuang
- State Key Laboratory of Natural Medicines, National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Yu
- State Key Laboratory of Natural Medicines, National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Lingjia Zhou
- State Key Laboratory of Natural Medicines, National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yong Li
- College of Life Science and Technology, Ningxia Polytechnic, Yingchuan, Ningxia 750021, China
| | - Chao Guo
- College of Life Science and Technology, Ningxia Polytechnic, Yingchuan, Ningxia 750021, China
| | - Chao Xu
- College of Life Science and Technology, Ningxia Polytechnic, Yingchuan, Ningxia 750021, China
| | - Linxian Li
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Hui Huang
- State Key Laboratory of Natural Medicines, National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, Nanjing 211198, China
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Jinsong Han
- State Key Laboratory of Natural Medicines, National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
4
|
Jemimah S, Abuhantash F, AlShehhi A. c-Triadem: A constrained, explainable deep learning model to identify novel biomarkers in Alzheimer's disease. PLoS One 2025; 20:e0320360. [PMID: 40228177 PMCID: PMC11996220 DOI: 10.1371/journal.pone.0320360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 02/17/2025] [Indexed: 04/16/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that requires early diagnosis for effective management. However, issues with currently available diagnostic biomarkers preclude early diagnosis, necessitating the development of alternative biomarkers and methods, such as blood-based diagnostics. We propose c-Triadem (constrained triple-input Alzheimer's disease model), a novel deep neural network to identify potential blood-based biomarkers for AD and predict mild cognitive impairment (MCI) and AD with high accuracy. The model utilizes genotyping data, gene expression data, and clinical information to predict the disease status of participants, i.e., cognitively normal (CN), MCI, or AD. The nodes of the neural network represent genes and their related pathways, and the edges represent known relationships among the genes and pathways. Simulated data validation further highlights the robustness of key features identified by SHapley Additive exPlanations (SHAP). We trained the model with blood genotyping data, microarray, and clinical features from the Alzheimer's Neuroimaging Disease Initiative (ADNI). We demonstrate that our model's performance is superior to previous models with an AUC of 97% and accuracy of 89%. We then identified the most influential genes and clinical features for prediction using SHapley Additive exPlanations (SHAP). Our SHAP analysis shows that CASP9, LCK, and SDC3 SNPs and PINK1, ATG5, and ubiquitin (UBB, UBC) expression have a higher impact on model performance. Our model has facilitated the identification of potential blood-based genetic markers of DNA damage response and mitophagy in affected regions of the brain. The model can be used for detection and biomarker identification in other related dementias.
Collapse
Affiliation(s)
- Sherlyn Jemimah
- Department of Biomedical Engineering and Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Ferial Abuhantash
- Department of Biomedical Engineering and Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Aamna AlShehhi
- Department of Biomedical Engineering and Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
5
|
Yates J, Van Allen EM. New horizons at the interface of artificial intelligence and translational cancer research. Cancer Cell 2025; 43:708-727. [PMID: 40233719 PMCID: PMC12007700 DOI: 10.1016/j.ccell.2025.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/04/2025] [Accepted: 03/12/2025] [Indexed: 04/17/2025]
Abstract
Artificial intelligence (AI) is increasingly being utilized in cancer research as a computational strategy for analyzing multiomics datasets. Advances in single-cell and spatial profiling technologies have contributed significantly to our understanding of tumor biology, and AI methodologies are now being applied to accelerate translational efforts, including target discovery, biomarker identification, patient stratification, and therapeutic response prediction. Despite these advancements, the integration of AI into clinical workflows remains limited, presenting both challenges and opportunities. This review discusses AI applications in multiomics analysis and translational oncology, emphasizing their role in advancing biological discoveries and informing clinical decision-making. Key areas of focus include cellular heterogeneity, tumor microenvironment interactions, and AI-aided diagnostics. Challenges such as reproducibility, interpretability of AI models, and clinical integration are explored, with attention to strategies for addressing these hurdles. Together, these developments underscore the potential of AI and multiomics to enhance precision oncology and contribute to advancements in cancer care.
Collapse
Affiliation(s)
- Josephine Yates
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland; ETH AI Center, ETH Zurich, Zurich, Switzerland; Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Medical Sciences, Harvard University, Boston, MA, USA; Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
6
|
Wu C, Andaloussi MA, Hormuth DA, Lima EABF, Lorenzo G, Stowers CE, Ravula S, Levac B, Dimakis AG, Tamir JI, Brock KK, Chung C, Yankeelov TE. A critical assessment of artificial intelligence in magnetic resonance imaging of cancer. NPJ IMAGING 2025; 3:15. [PMID: 40226507 PMCID: PMC11981920 DOI: 10.1038/s44303-025-00076-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 03/17/2025] [Indexed: 04/15/2025]
Abstract
Given the enormous output and pace of development of artificial intelligence (AI) methods in medical imaging, it can be challenging to identify the true success stories to determine the state-of-the-art of the field. This report seeks to provide the magnetic resonance imaging (MRI) community with an initial guide into the major areas in which the methods of AI are contributing to MRI in oncology. After a general introduction to artificial intelligence, we proceed to discuss the successes and current limitations of AI in MRI when used for image acquisition, reconstruction, registration, and segmentation, as well as its utility for assisting in diagnostic and prognostic settings. Within each section, we attempt to present a balanced summary by first presenting common techniques, state of readiness, current clinical needs, and barriers to practical deployment in the clinical setting. We conclude by presenting areas in which new advances must be realized to address questions regarding generalizability, quality assurance and control, and uncertainty quantification when applying MRI to cancer to maintain patient safety and practical utility.
Collapse
Affiliation(s)
- Chengyue Wu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
| | | | - David A. Hormuth
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX USA
| | - Ernesto A. B. F. Lima
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
- Texas Advanced Computing Center, The University of Texas at Austin, Austin, TX USA
| | - Guillermo Lorenzo
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
- Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Casey E. Stowers
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
| | - Sriram Ravula
- Chandra Family Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX USA
| | - Brett Levac
- Chandra Family Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX USA
| | - Alexandros G. Dimakis
- Chandra Family Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX USA
| | - Jonathan I. Tamir
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
- Chandra Family Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX USA
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX USA
| | - Kristy K. Brock
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Caroline Chung
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Thomas E. Yankeelov
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX USA
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX USA
- Department of Oncology, The University of Texas at Austin, Austin, TX USA
| |
Collapse
|
7
|
Su J, He K, Li Y, Tu J, Chen X. Soft Materials and Devices Enabling Sensorimotor Functions in Soft Robots. Chem Rev 2025. [PMID: 40163535 DOI: 10.1021/acs.chemrev.4c00906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Sensorimotor functions, the seamless integration of sensing, decision-making, and actuation, are fundamental for robots to interact with their environments. Inspired by biological systems, the incorporation of soft materials and devices into robotics holds significant promise for enhancing these functions. However, current robotics systems often lack the autonomy and intelligence observed in nature due to limited sensorimotor integration, particularly in flexible sensing and actuation. As the field progresses toward soft, flexible, and stretchable materials, developing such materials and devices becomes increasingly critical for advanced robotics. Despite rapid advancements individually in soft materials and flexible devices, their combined applications to enable sensorimotor capabilities in robots are emerging. This review addresses this emerging field by providing a comprehensive overview of soft materials and devices that enable sensorimotor functions in robots. We delve into the latest development in soft sensing technologies, actuation mechanism, structural designs, and fabrication techniques. Additionally, we explore strategies for sensorimotor control, the integration of artificial intelligence (AI), and practical application across various domains such as healthcare, augmented and virtual reality, and exploration. By drawing parallels with biological systems, this review aims to guide future research and development in soft robots, ultimately enhancing the autonomy and adaptability of robots in unstructured environments.
Collapse
Affiliation(s)
- Jiangtao Su
- Innovative Centre for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Ke He
- Innovative Centre for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Yanzhen Li
- Innovative Centre for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Jiaqi Tu
- Innovative Centre for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Xiaodong Chen
- Innovative Centre for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| |
Collapse
|
8
|
Jiang Y, Aton M, Zhu Q, Lu YY. Modeling microbiome-trait associations with taxonomy-adaptive neural networks. MICROBIOME 2025; 13:87. [PMID: 40158141 PMCID: PMC11954268 DOI: 10.1186/s40168-025-02080-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
The human microbiome, a complex ecosystem of microorganisms inhabiting the body, plays a critical role in human health. Investigating its association with host traits is essential for understanding its impact on various diseases. Although shotgun metagenomic sequencing technologies have produced vast amounts of microbiome data, analyzing such data is highly challenging due to its sparsity, noisiness, and high feature dimensionality. Here, we develop MIOSTONE, an accurate and interpretable neural network model for microbiome-disease association that simulates a real taxonomy by encoding the relationships among microbial features. The taxonomy-encoding architecture provides a natural bridge from variations in microbial taxa abundance to variations in traits, encompassing increasingly coarse scales from species to domains. MIOSTONE has the ability to determine whether taxa within the corresponding taxonomic group provide a better explanation in a data-driven manner. MIOSTONE serves as an effective predictive model, as it not only accurately predicts microbiome-trait associations across extensive simulated and real datasets but also offers interpretability for scientific discovery. Both attributes are crucial for facilitating in silico investigations into the biological mechanisms underlying such associations among microbial taxa. Video Abstract.
Collapse
Affiliation(s)
- Yifan Jiang
- Cheriton School of Computer Science, University of Waterloo, Waterloo, Ontario, Canada
| | - Matthew Aton
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Qiyun Zhu
- School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| | - Yang Young Lu
- Cheriton School of Computer Science, University of Waterloo, Waterloo, Ontario, Canada.
| |
Collapse
|
9
|
Thapa K, Kinali M, Pei S, Luna A, Babur Ö. Strategies to include prior knowledge in omics analysis with deep neural networks. PATTERNS (NEW YORK, N.Y.) 2025; 6:101203. [PMID: 40182174 PMCID: PMC11963003 DOI: 10.1016/j.patter.2025.101203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
High-throughput molecular profiling technologies have revolutionized molecular biology research in the past decades. One important use of molecular data is to make predictions of phenotypes and other features of the organisms using machine learning algorithms. Deep learning models have become increasingly popular for this task due to their ability to learn complex non-linear patterns. Applying deep learning to molecular profiles, however, is challenging due to the very high dimensionality of the data and relatively small sample sizes, causing models to overfit. A solution is to incorporate biological prior knowledge to guide the learning algorithm for processing the functionally related input together. This helps regularize the models and improve their generalizability and interpretability. Here, we describe three major strategies proposed to use prior knowledge in deep learning models to make predictions based on molecular profiles. We review the related deep learning architectures, including the major ideas in relatively new graph neural networks.
Collapse
Affiliation(s)
- Kisan Thapa
- Computer Science Department, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, MA 02125, USA
| | - Meric Kinali
- Computer Science Department, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, MA 02125, USA
| | - Shichao Pei
- Computer Science Department, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, MA 02125, USA
| | - Augustin Luna
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bathesda, MD 20892, USA
- Computational Biology Branch, National Library of Medicine, NIH, 9000 Rockville Pike, Bathesda, MD 20892, USA
| | - Özgün Babur
- Computer Science Department, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, MA 02125, USA
| |
Collapse
|
10
|
Guo Y, Sun X, Li L, Shi Y, Cheng W, Pan L. Deep-Learning-Based Analysis of Electronic Skin Sensing Data. SENSORS (BASEL, SWITZERLAND) 2025; 25:1615. [PMID: 40096464 PMCID: PMC11902811 DOI: 10.3390/s25051615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
E-skin is an integrated electronic system that can mimic the perceptual ability of human skin. Traditional analysis methods struggle to handle complex e-skin data, which include time series and multiple patterns, especially when dealing with intricate signals and real-time responses. Recently, deep learning techniques, such as the convolutional neural network, recurrent neural network, and transformer methods, provide effective solutions that can automatically extract data features and recognize patterns, significantly improving the analysis of e-skin data. Deep learning is not only capable of handling multimodal data but can also provide real-time response and personalized predictions in dynamic environments. Nevertheless, problems such as insufficient data annotation and high demand for computational resources still limit the application of e-skin. Optimizing deep learning algorithms, improving computational efficiency, and exploring hardware-algorithm co-designing will be the key to future development. This review aims to present the deep learning techniques applied in e-skin and provide inspiration for subsequent researchers. We first summarize the sources and characteristics of e-skin data and review the deep learning models applicable to e-skin data and their applications in data analysis. Additionally, we discuss the use of deep learning in e-skin, particularly in health monitoring and human-machine interactions, and we explore the current challenges and future development directions.
Collapse
Affiliation(s)
| | | | | | - Yi Shi
- Collaborative Innovation Center of Advanced Microstructures, School of Electronic Science and Engineering, Nanjing University, Nanjing 210093, China; (Y.G.); (X.S.); (L.L.)
| | - Wen Cheng
- Collaborative Innovation Center of Advanced Microstructures, School of Electronic Science and Engineering, Nanjing University, Nanjing 210093, China; (Y.G.); (X.S.); (L.L.)
| | - Lijia Pan
- Collaborative Innovation Center of Advanced Microstructures, School of Electronic Science and Engineering, Nanjing University, Nanjing 210093, China; (Y.G.); (X.S.); (L.L.)
| |
Collapse
|
11
|
Selby DA, Sprang M, Ewald J, Vollmer SJ. Beyond the black box with biologically informed neural networks. Nat Rev Genet 2025:10.1038/s41576-025-00826-1. [PMID: 40038452 DOI: 10.1038/s41576-025-00826-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Affiliation(s)
- David A Selby
- Data Science and its Applications, German Research Center for Artificial Intelligence (DFKI), Kaiserslautern, Germany.
| | - Maximilian Sprang
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jan Ewald
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI) Dresden/Leipzig, Leipzig University, Leipzig, Germany
| | - Sebastian J Vollmer
- Data Science and its Applications, German Research Center for Artificial Intelligence (DFKI), Kaiserslautern, Germany
- University of Kaiserslautern-Landau, Kaiserslautern, Germany
| |
Collapse
|
12
|
Ligero M, El Nahhas OSM, Aldea M, Kather JN. Artificial intelligence-based biomarkers for treatment decisions in oncology. Trends Cancer 2025; 11:232-244. [PMID: 39814650 DOI: 10.1016/j.trecan.2024.12.001] [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: 07/02/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Abstract
The development of new therapeutic strategies such as immune checkpoint inhibitors (ICIs) and targeted therapies has increased the complexity of the treatment landscape for solid tumors. At the current rate of annual FDA approvals, the potential treatment options could increase by tenfold over the next 5 years. The cost of personalized medicine technologies limits its accessibility, thus increasing socioeconomic disparities in the treated population. In this review we describe artificial intelligence (AI)-based solutions - including deep learning (DL) methods for routine medical imaging and large language models (LLMs) for electronic health records (EHRs) - to support cancer treatment decisions with cost-effective biomarkers. We address the current limitations of these technologies and propose the next steps towards their adoption in routine clinical practice.
Collapse
Affiliation(s)
- Marta Ligero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany
| | - Omar S M El Nahhas
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany
| | - Mihaela Aldea
- Department of Cancer Medicine, Institut Gustave Roussy, Université Paris-Saclay, F-94805, Villejuif, France; Thoracic Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany; Department of Medicine I, University Hospital Dresden, Dresden, Germany; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
13
|
Yoo SK, Fitzgerald CW, Cho BA, Fitzgerald BG, Han C, Koh ES, Pandey A, Sfreddo H, Crowley F, Korostin MR, Debnath N, Leyfman Y, Valero C, Lee M, Vos JL, Lee AS, Zhao K, Lam S, Olumuyide E, Kuo F, Wilson EA, Hamon P, Hennequin C, Saffern M, Vuong L, Hakimi AA, Brown B, Merad M, Gnjatic S, Bhardwaj N, Galsky MD, Schadt EE, Samstein RM, Marron TU, Gönen M, Morris LGT, Chowell D. Prediction of checkpoint inhibitor immunotherapy efficacy for cancer using routine blood tests and clinical data. Nat Med 2025; 31:869-880. [PMID: 39762425 PMCID: PMC11922749 DOI: 10.1038/s41591-024-03398-5] [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: 03/25/2024] [Accepted: 11/01/2024] [Indexed: 01/25/2025]
Abstract
Predicting whether a patient with cancer will benefit from immune checkpoint inhibitors (ICIs) without resorting to advanced genomic or immunologic assays is an important clinical need. To address this, we developed and evaluated SCORPIO, a machine learning system that utilizes routine blood tests (complete blood count and comprehensive metabolic profile) alongside clinical characteristics from 9,745 ICI-treated patients across 21 cancer types. SCORPIO was trained on data from 1,628 patients across 17 cancer types from Memorial Sloan Kettering Cancer Center. In two internal test sets comprising 2,511 patients across 19 cancer types, SCORPIO achieved median time-dependent area under the receiver operating characteristic curve (AUC(t)) values of 0.763 and 0.759 for predicting overall survival at 6, 12, 18, 24 and 30 months, outperforming tumor mutational burden (TMB), which showed median AUC(t) values of 0.503 and 0.543. Additionally, SCORPIO demonstrated superior predictive performance for predicting clinical benefit (tumor response or prolonged stability), with AUC values of 0.714 and 0.641, compared to TMB (AUC = 0.546 and 0.573). External validation was performed using 10 global phase 3 trials (4,447 patients across 6 cancer types) and a real-world cohort from the Mount Sinai Health System (1,159 patients across 18 cancer types). In these external cohorts, SCORPIO maintained robust performance in predicting ICI outcomes, surpassing programmed death-ligand 1 immunostaining. These findings underscore SCORPIO's reliability and adaptability, highlighting its potential to predict patient outcomes with ICI therapy across diverse cancer types and healthcare settings.
Collapse
Affiliation(s)
- Seong-Keun Yoo
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Conall W Fitzgerald
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Byuri Angela Cho
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bailey G Fitzgerald
- Department of Medicine, Thoracic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Catherine Han
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth S Koh
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Abhinav Pandey
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hannah Sfreddo
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fionnuala Crowley
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brookdale Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Neha Debnath
- Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside and West, New York, NY, USA
| | - Yan Leyfman
- Internal Medicine, Icahn School of Medicine at Mount Sinai South Nassau, Rockville Centre, NY, USA
| | - Cristina Valero
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark Lee
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joris L Vos
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew Sangho Lee
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karena Zhao
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stanley Lam
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ezekiel Olumuyide
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fengshen Kuo
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eric A Wilson
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pauline Hamon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Clotilde Hennequin
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Saffern
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lynda Vuong
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Ari Hakimi
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brian Brown
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nina Bhardwaj
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Galsky
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Robert M Samstein
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas U Marron
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Early Phase Trials Unit, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luc G T Morris
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Diego Chowell
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
14
|
Nilsson A, Meimetis N, Lauffenburger DA. Towards an interpretable deep learning model of cancer. NPJ Precis Oncol 2025; 9:46. [PMID: 39948231 PMCID: PMC11825879 DOI: 10.1038/s41698-025-00822-y] [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: 09/26/2024] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Cancer is a manifestation of dysfunctional cell states. It emerges from an interplay of intrinsic and extrinsic factors that disrupt cellular dynamics, including genetic and epigenetic alterations, as well as the tumor microenvironment. This complexity can make it challenging to infer molecular causes for treating the disease. This may be addressed by system-wide computer models of cells, as they allow rapid generation and testing of hypotheses that would be too slow or impossible to perform in the laboratory and clinic. However, so far, such models have been impeded by both experimental and computational limitations. In this perspective, we argue that they can now be achieved using deep learning algorithms to integrate omics data and prior knowledge of molecular networks. Such models would have many applications in precision oncology, e.g., for identifying drug targets and biomarkers, predicting resistance mechanisms and toxicity effects of drugs, or simulating cell-cell interactions in the microenvironment.
Collapse
Affiliation(s)
- Avlant Nilsson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg, Sweden
- Department of Cell and Molecular Biology, SciLifeLab, Karolinska Institutet, Stockholm, Sweden
| | - Nikolaos Meimetis
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
15
|
Kennedy L, Sandhu JK, Harper ME, Cuperlovic-Culf M. A hybrid machine learning framework for functional annotation of mitochondrial glutathione transport and metabolism proteins in cancers. BMC Bioinformatics 2025; 26:48. [PMID: 39934670 PMCID: PMC11817629 DOI: 10.1186/s12859-025-06051-1] [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: 03/18/2024] [Accepted: 01/15/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Alterations of metabolism, including changes in mitochondrial metabolism as well as glutathione (GSH) metabolism are a well appreciated hallmark of many cancers. Mitochondrial GSH (mGSH) transport is a poorly characterized aspect of GSH metabolism, which we investigate in the context of cancer. Existing functional annotation approaches from machine (ML) or deep learning (DL) models based only on protein sequences, were unable to annotate functions in biological contexts. RESULTS We develop a flexible ML framework for functional annotation from diverse feature data. This hybrid ML framework leverages cancer cell line multi-omics data and other biological knowledge data as features, to uncover potential genes involved in mGSH metabolism and membrane transport in cancers. This framework achieves strong performance across functional annotation tasks and several cell line and primary tumor cancer samples. For our application, classification models predict the known mGSH transporter SLC25A39 but not SLC25A40 as being highly probably related to mGSH metabolism in cancers. SLC25A10, SLC25A50, and orphan SLC25A24, SLC25A43 are predicted to be associated with mGSH metabolism in multiple biological contexts and structural analysis of these proteins reveal similarities in potential substrate binding regions to the binding residues of SLC25A39. CONCLUSION These findings have implications for a better understanding of cancer cell metabolism and novel therapeutic targets with respect to GSH metabolism through potential novel functional annotations of genes. The hybrid ML framework proposed here can be applied to other biological function classifications or multi-omics datasets to generate hypotheses in various biological contexts. Code and a tutorial for generating models and predictions in this framework are available at: https://github.com/lkenn012/mGSH_cancerClassifiers .
Collapse
Affiliation(s)
- Luke Kennedy
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Jagdeep K Sandhu
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Bldg M54, Ottawa, ON, K1A 0R6, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| | - Miroslava Cuperlovic-Culf
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
- Digital Technologies Research Centre, National Research Council Canada, 1200 Montreal Road, Bldg M50, Ottawa, ON, K1A 0R6, Canada.
| |
Collapse
|
16
|
Singhal A, Zhao X, Wall P, So E, Calderini G, Partin A, Koussa N, Vasanthakumari P, Narykov O, Zhu Y, Jones SE, Abbas-Aghababazadeh F, Nair SK, Bélisle-Pipon JC, Jayaram A, Parker BA, Yeung KT, Griffiths JI, Weil R, Nath A, Haibe-Kains B, Ideker T. The Hallmarks of Predictive Oncology. Cancer Discov 2025; 15:271-285. [PMID: 39760657 PMCID: PMC11969157 DOI: 10.1158/2159-8290.cd-24-0760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/30/2024] [Accepted: 10/16/2024] [Indexed: 01/07/2025]
Abstract
SIGNIFICANCE As the field of artificial intelligence evolves rapidly, these hallmarks are intended to capture fundamental, complementary concepts necessary for the progress and timely adoption of predictive modeling in precision oncology. Through these hallmarks, we hope to establish standards and guidelines that enable the symbiotic development of artificial intelligence and precision oncology.
Collapse
Affiliation(s)
- Akshat Singhal
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Xiaoyu Zhao
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Patrick Wall
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Emily So
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Guido Calderini
- Faculty of Health Science, Simon Fraser University, Burnaby, BC, Canada
- École de santé publique, Université de Montréal, Montréal, QC, Canada
| | - Alexander Partin
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
| | - Natasha Koussa
- Cancer Data Science Initiatives, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Oleksandr Narykov
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
| | - Yitan Zhu
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
| | - Sara E. Jones
- Cancer Data Science Initiatives, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | | | | | | - Barbara A. Parker
- Moores Cancer Center, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Kay T. Yeung
- Moores Cancer Center, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jason I. Griffiths
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute, City of Hope National Medical Center, Monrovia, CA, USA
| | - Ryan Weil
- Cancer Data Science Initiatives, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Aritro Nath
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute, City of Hope National Medical Center, Monrovia, CA, USA
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Medical Biophysics, University of Toronto, Toronto, Canada
- Vector Institute for Artificial Intelligence, Toronto, Canada
- Department of Biostatistics, Dalla Lana School of Public Health, Toronto, Canada
| | - Trey Ideker
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
17
|
Azadinejad H, Farhadi Rad M, Shariftabrizi A, Rahmim A, Abdollahi H. Optimizing Cancer Treatment: Exploring the Role of AI in Radioimmunotherapy. Diagnostics (Basel) 2025; 15:397. [PMID: 39941326 PMCID: PMC11816985 DOI: 10.3390/diagnostics15030397] [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: 12/10/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Radioimmunotherapy (RIT) is a novel cancer treatment that combines radiotherapy and immunotherapy to precisely target tumor antigens using monoclonal antibodies conjugated with radioactive isotopes. This approach offers personalized, systemic, and durable treatment, making it effective in cancers resistant to conventional therapies. Advances in artificial intelligence (AI) present opportunities to enhance RIT by improving precision, efficiency, and personalization. AI plays a critical role in patient selection, treatment planning, dosimetry, and response assessment, while also contributing to drug design and tumor classification. This review explores the integration of AI into RIT, emphasizing its potential to optimize the entire treatment process and advance personalized cancer care.
Collapse
Affiliation(s)
- Hossein Azadinejad
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah 6714869914, Iran;
| | - Mohammad Farhadi Rad
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Ahmad Shariftabrizi
- Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA;
| | - Arman Rahmim
- Department of Radiology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 0B4, Canada
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Hamid Abdollahi
- Department of Radiology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 0B4, Canada
| |
Collapse
|
18
|
Xi X, Li J, Jia J, Meng Q, Li C, Wang X, Wei L, Zhang X. A mechanism-informed deep neural network enables prioritization of regulators that drive cell state transitions. Nat Commun 2025; 16:1284. [PMID: 39900922 PMCID: PMC11790924 DOI: 10.1038/s41467-025-56475-9] [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: 06/02/2024] [Accepted: 01/15/2025] [Indexed: 02/05/2025] Open
Abstract
Cells are regulated at multiple levels, from regulations of individual genes to interactions across multiple genes. Some recent neural network models can connect molecular changes to cellular phenotypes, but their design lacks modeling of regulatory mechanisms, limiting the decoding of regulations behind key cellular events, such as cell state transitions. Here, we present regX, a deep neural network incorporating both gene-level regulation and gene-gene interaction mechanisms, which enables prioritizing potential driver regulators of cell state transitions and providing mechanistic interpretations. Applied to single-cell multi-omics data on type 2 diabetes and hair follicle development, regX reliably prioritizes key transcription factors and candidate cis-regulatory elements that drive cell state transitions. Some regulators reveal potential new therapeutic targets, drug repurposing possibilities, and putative causal single nucleotide polymorphisms. This method to analyze single-cell multi-omics data demonstrates how the interpretable design of neural networks can better decode biological systems.
Collapse
Affiliation(s)
- Xi Xi
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Jiaqi Li
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Jinmeng Jia
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Qiuchen Meng
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Chen Li
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Xiaowo Wang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Lei Wei
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Xuegong Zhang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China.
- School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
19
|
Zhou L, Li J, Tan W. M-NET: Transforming Single Nucleotide Variations Into Patient Feature Images for the Prediction of Prostate Cancer Metastasis and Identification of Significant Pathways. IEEE J Biomed Health Inform 2025; 29:1199-1208. [PMID: 39509309 DOI: 10.1109/jbhi.2024.3493618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
High-performance prediction of prostate cancer metastasis based on single nucleotide variations remains a challenge. Therefore, we developed a novel biologically informed deep learning framework, named M-NET, for the prediction of prostate cancer metastasis. Within the framework, we transformed single nucleotide variations into patient feature images that are optimal for fitting convolutional neural networks. Moreover, we identified significant pathways associated with the metastatic status. The experimental results showed that M-NET significantly outperformed other comparison methods based on single nucleotide variations, achieving improvements in accuracy, precision, recall, F1-score, area under the receiver operating characteristics curve, and area under the precision-recall curve by 6.3%, 8.4%, 5.1%, 0.070, 0.041, and 0.026, respectively. Furthermore, M-NET identified some important pathways associated with the metastatic status, such as signaling by the hedgehog pathway. In summary, compared with other comparative methods, M-NET exhibited a better performance in the prediction of prostate cancer metastasis.
Collapse
|
20
|
Miyahira AK, Sharifi M, Chesner LN, El-Kenawi A, Haas R, Sena LA, Tewari AK, Pienta KJ, Soule HR. Personalized Medicine: Leave no Patient Behind; Report From the 2024 Coffey-Holden Prostate Cancer Academy Meeting. Prostate 2025; 85:211-226. [PMID: 39604057 DOI: 10.1002/pros.24826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024]
Abstract
INTRODUCTION The 11th Annual 2024 Coffey - Holden Prostate Cancer Academy (CHPCA) Meeting, was themed "Personalized Medicine: Leave No Patient Behind," and was held from June 20 to 23, 2024 at the University of California, Los Angeles, Luskin Conference Center, in Los Angeles, CA. METHODS The CHPCA Meeting is an academy-styled annual conference organized by the Prostate Cancer Foundation, to focus discussion on the most critical emerging research that have the greatest potential to advance knowledge of prostate cancer biology and treatment. The 2024 CHPCA Meeting was attended by 75 academic investigators and included 37 talks across 8 sessions. RESULTS The meeting sessions focused on: novel human, mouse and systems biology research models, novel immunotherapies for prostate cancer, efforts to overcome treatment resistance, the role of metabolism and diet in prostate cancer biology and as a therapeutic target, mechanisms that drive differentiation into neuroendocrine cancer subtypes, the evolving prostate cancer epigenome in disease progression and treatment resistance, and machine learning and advanced computational approaches for precision oncology. DISCUSSION This article summarizes the presentations and discussions from the 2024 CHPCA Meeting. We hope that sharing this knowledge will inspire and accelerate research into new discoveries and solutions for prostate cancer.
Collapse
Affiliation(s)
- Andrea K Miyahira
- Department of Science, Prostate Cancer Foundation, Santa Monica, California, USA
| | - Marina Sharifi
- Department of Medicine and Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lisa N Chesner
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California, USA
| | - Asmaa El-Kenawi
- Department of Urology, Indiana University School of Medicine, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana, USA
| | - Roni Haas
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California, USA
| | - Laura A Sena
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Alok K Tewari
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kenneth J Pienta
- The James Buchanan Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Howard R Soule
- Department of Science, Prostate Cancer Foundation, Santa Monica, California, USA
| |
Collapse
|
21
|
Li H, Zeng J, Snyder MP, Zhang S. Modeling gene interactions in polygenic prediction via geometric deep learning. Genome Res 2025; 35:178-187. [PMID: 39562137 PMCID: PMC11789630 DOI: 10.1101/gr.279694.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
Polygenic risk score (PRS) is a widely used approach for predicting individuals' genetic risk of complex diseases, playing a pivotal role in advancing precision medicine. Traditional PRS methods, predominantly following a linear structure, often fall short in capturing the intricate relationships between genotype and phenotype. In this study, we present PRS-Net, an interpretable geometric deep learning-based framework that effectively models the nonlinearity of biological systems for enhanced disease prediction and biological discovery. PRS-Net begins by deconvoluting the genome-wide PRS at the single-gene resolution and then explicitly encapsulates gene-gene interactions leveraging a graph neural network (GNN) for genetic risk prediction, enabling a systematic characterization of molecular interplay underpinning diseases. An attentive readout module is introduced to facilitate model interpretation. Extensive tests across multiple complex traits and diseases demonstrate the superior prediction performance of PRS-Net compared with a wide range of conventional PRS methods. The interpretability of PRS-Net further enhances the identification of disease-relevant genes and gene programs. PRS-Net provides a potent tool for concurrent genetic risk prediction and biological discovery for complex diseases.
Collapse
Affiliation(s)
- Han Li
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, 300071, China
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, 100084, China
| | - Jianyang Zeng
- School of Engineering, Research Center for Industries of the Future, Westlake University, Hangzhou, 310030, Zhejiang, China;
| | - Michael P Snyder
- Department of Genetics, Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, California 94304, USA;
| | - Sai Zhang
- Department of Epidemiology, University of Florida, Gainesville, Florida 32603, USA;
- Departments of Biostatistics & Biomedical Engineering, UF Genetics Institute, University of Florida, Gainesville, Florida 32603, USA
| |
Collapse
|
22
|
Wu T, Zhao L, Ren M, He S, Zhang L, Fang M, Wang B. Small-Sample Learning for Next-Generation Human Health Risk Assessment: Harnessing AI, Exposome Data, and Systems Biology. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:5-10. [PMID: 39745662 DOI: 10.1021/acs.est.4c11832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Affiliation(s)
- Tianxiang Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of PR China, Beijing 100191, China
| | - Lu Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of PR China, Beijing 100191, China
| | - Mengyuan Ren
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of PR China, Beijing 100191, China
| | - Song He
- Department of Bioinformatics, Academy of Military Medical Sciences, Beijing 100850, China
| | - Le Zhang
- School of Information and Communication Engineering, University of Electronic Science and Technology of China (UESTC), Chengdu 611731, China
| | - Mingliang Fang
- Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China
| | - Bin Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of PR China, Beijing 100191, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China
- Laboratory for Earth Surface Processes, College of Urban and Environmental Science, Peking University, Beijing 100871, China
| |
Collapse
|
23
|
Liu HM, Liu ZF, Li Z, Yu C, Hu PC, Liu QF, Shi TG. Genome-wide association study on color-image-based convolutional neural networks. PeerJ 2025; 13:e18822. [PMID: 39822975 PMCID: PMC11737327 DOI: 10.7717/peerj.18822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 12/16/2024] [Indexed: 01/19/2025] Open
Abstract
Background Convolutional neural networks have excellent modeling abilities to complex large-scale datasets and have been applied to genomics. It requires converting genotype data to image format when employing convolutional neural networks to genome-wide association studies. Existing studies converting the data into grayscale images have shown promising. However, the grayscale image may cause the loss of information of the genotype data. Methods In order to make full use of the information, we proposed a new method, color-image-based convolutional neural networks, by converting the data into color images. Results The experiments on simulation and real data show that our method outperforms the existing methods proposed by Yue and Chen for converting data into grayscale images, in which the model accuracy is improved by an average of 7.61%, and the ratio of disease risk genes is increased by an average of 18.91%. The new method has better robustness and generalized performance.
Collapse
Affiliation(s)
- Han-Ming Liu
- School of Mathematics and Computer Science, Gannan Normal University, Ganzhou, China
| | | | - Zi Li
- School of Mathematics and Computer Science, Gannan Normal University, Ganzhou, China
| | - Cong Yu
- School of Mathematics and Computer Science, Gannan Normal University, Ganzhou, China
| | - Peng-Cheng Hu
- School of Mathematics and Computer Science, Gannan Normal University, Ganzhou, China
| | - Qi-Feng Liu
- School of Mathematics and Computer Science, Gannan Normal University, Ganzhou, China
| | - Tai-Gui Shi
- School of Mathematics and Computer Science, Gannan Normal University, Ganzhou, China
| |
Collapse
|
24
|
Rai HM, Yoo J, Dashkevych S. Transformative Advances in AI for Precise Cancer Detection: A Comprehensive Review of Non-Invasive Techniques. ARCHIVES OF COMPUTATIONAL METHODS IN ENGINEERING 2025. [DOI: 10.1007/s11831-024-10219-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 12/07/2024] [Indexed: 03/02/2025]
|
25
|
Zeng S, Adusumilli T, Awan SZ, Immadi MS, Xu D, Joshi T. G2PDeep-v2: a web-based deep-learning framework for phenotype prediction and biomarker discovery for all organisms using multi-omics data. RESEARCH SQUARE 2025:rs.3.rs-5776937. [PMID: 39866874 PMCID: PMC11760241 DOI: 10.21203/rs.3.rs-5776937/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The G2PDeep-v2 server is a web-based platform powered by deep learning, for phenotype prediction and markers discovery from multi-omics data in any organisms including humans, plants, animals, and viruses. The server provides multiple services for researchers to create deep-learning models through an interactive interface and train these models using an automated hyperparameter tuning algorithm on high-performance computing resources. Users can visualize the results of phenotype and markers predictions and perform Gene Set Enrichment Analysis for the significant markers to provide insights into the molecular mechanisms underlying complex diseases, conditions and other biological phenotypes being studied. The G2PDeep-v2 server is publicly available at https://g2pdeep.org/ and can be utilized for all organisms.
Collapse
|
26
|
Unger M, Loeffler CML, Žigutytė L, Sainath S, Lenz T, Vibert J, Mock A, Fröhling S, Graham TA, Carrero ZI, Kather JN. Deep Learning for Biomarker Discovery in Cancer Genomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631471. [PMID: 39829845 PMCID: PMC11741323 DOI: 10.1101/2025.01.06.631471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Background Genomic data is essential for clinical decision-making in precision oncology. Bioinformatic algorithms are widely used to analyze next-generation sequencing (NGS) data, but they face two major challenges. First, these pipelines are highly complex, involving multiple steps and the integration of various tools. Second, they generate features that are human-interpretable but often result in information loss by focusing only on predefined genetic properties. This limitation restricts the full potential of NGS data in biomarker extraction and slows the discovery of new biomarkers in precision oncology. Methods We propose an end-to-end deep learning (DL) approach for analyzing NGS data. Specifically, we developed a multiple instance learning DL framework that integrates somatic mutation sequences to predict two compound biomarkers: microsatellite instability (MSI) and homologous recombination deficiency (HRD). To achieve this, we utilized data from 3,184 cancer patients obtained from two public databases: The Cancer Genome Atlas (TCGA) and the Clinical Proteome Tumor Analysis Consortium (CPTAC). Results Our proposed deep learning method demonstrated high accuracy in identifying clinically relevant biomarkers. For predicting MSI status, the model achieved an accuracy of 0.98, a sensitivity of 0.95, and a specificity of 1.00 on an external validation cohort. For predicting HRD status, the model achieved an accuracy of 0.80, a sensitivity of 0.75, and a specificity of 0.86. Furthermore, the deep learning approach significantly outperformed traditional machine learning methods in both tasks (MSI accuracy, p-value = 5.11×10-18; HRD accuracy, p-value = 1.07×10-10). Using explainability techniques, we demonstrated that the model's predictions are based on biologically meaningful features, aligning with key DNA damage repair mutation signatures. Conclusion We demonstrate that deep learning can identify patterns in unfiltered somatic mutations without the need for manual feature extraction. This approach enhances the detection of actionable targets and paves the way for developing NGS-based biomarkers using minimally processed data.
Collapse
Affiliation(s)
- Michaela Unger
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
| | - Chiara M L Loeffler
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
- Medical Department 1, University Hospital and Faculty of Medicine Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Laura Žigutytė
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
| | - Srividhya Sainath
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
| | - Tim Lenz
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
| | - Julien Vibert
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - Andreas Mock
- Institute of Pathology, Ludwig-Maximilians-University München, Munich, Germany
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Fröhling
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany
- Division of Translational Precision Medicine, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Trevor A Graham
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK
| | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
- Medical Department 1, University Hospital and Faculty of Medicine Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
27
|
Lama R, Fose JM, Martín D, Muñoz IG, Wang ES, Sung PJ, Chemler SR, Wang X. Novel Inhibitors for MDM2-MDM4 E3 Ligase Potently Induce p53-Indepedent Apoptosis in Drug-Resistant Leukemic Cells. Molecules 2025; 30:186. [PMID: 39795242 PMCID: PMC11722259 DOI: 10.3390/molecules30010186] [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: 12/05/2024] [Revised: 12/31/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025] Open
Abstract
MDM2 and MDM4 are major negative regulators of tumor suppressor p53. Beyond regulating p53, MDM2 possesses p53-independent activity in promoting cell cycle progression and tumorigenesis via its RING domain ubiquitin E3 ligase activity. MDM2 and MDM4 form heterodimer polyubiquitin E3 ligases via their RING domain interaction. Inhibitors disrupting p53 interaction with MDM2/MDM4 are in clinical trials in patients bearing wild-type p53 cancers. However, these inhibitors are not designed to work for p53-null/mutant cancer cells. Owing to the importance of the E3 ligase of MDM2 in its p53-independent oncogenic activity, inhibitors targeting the E3 ligase activity of MDM2-MDM4 are desirable for p53-mutant cancer cells. Here, we report the development of such inhibitors with pro-apoptotic activity in p53-null leukemic cells. Among analogues of MDM2-MDM4 E3 ligase inhibitors, we initially identified MMRi36 as a potent pro-apoptotic compound in p53-null leukemic cells with acquired drug resistance. MMRi36 acts as an activator of MDM2-MDM4 E3 ligase by stabilizing MDM2-MDM4 heterodimers and promotes MDM2/MDM4 degradation in cells. Interestingly, replacement of the sulfur in 1,3,4-thiadiazole MMRi36 with a carbon led to identification of pyrazole MMRi36C that dissociates the MDM2-MDM4 RING heterodimers, inhibits the E3 ligase activity of the complex, and induces p53 protein accumulation, but retains the p53-independent pro-apoptotic activity. A brief SAR study identified a fluorine derivative of MMRi36C with improved pro-apoptotic activity. This study discovered a novel class of compound that targets MDM2-MDM4 ubiquitin E3 ligase activity for apoptosis induction in p53-mutant cancer cells.
Collapse
Affiliation(s)
- Rati Lama
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (R.L.); (P.J.S.)
| | - Joseph M. Fose
- Department of Chemistry, University at Buffalo, State University of New York, Buffalo, NY 14260, USA; (J.M.F.); (S.R.C.)
| | - Diana Martín
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), 28009 Madrid, Spain; (D.M.); (I.G.M.)
| | - Inés G. Muñoz
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), 28009 Madrid, Spain; (D.M.); (I.G.M.)
| | - Eunice S. Wang
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Pamela J. Sung
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (R.L.); (P.J.S.)
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Sherry R. Chemler
- Department of Chemistry, University at Buffalo, State University of New York, Buffalo, NY 14260, USA; (J.M.F.); (S.R.C.)
| | - Xinjiang Wang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (R.L.); (P.J.S.)
| |
Collapse
|
28
|
Anh NK, Thu NQ, Tien NTN, Long NP, Nguyen HT. Advancements in Mass Spectrometry-Based Targeted Metabolomics and Lipidomics: Implications for Clinical Research. Molecules 2024; 29:5934. [PMID: 39770023 PMCID: PMC11677340 DOI: 10.3390/molecules29245934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/30/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Targeted metabolomics and lipidomics are increasingly utilized in clinical research, providing quantitative and comprehensive assessments of metabolic profiles that underlie physiological and pathological mechanisms. These approaches enable the identification of critical metabolites and metabolic alterations essential for accurate diagnosis and precision treatment. Mass spectrometry, in combination with various separation techniques, offers a highly sensitive and specific platform for implementing targeted metabolomics and lipidomics in clinical settings. Nevertheless, challenges persist in areas such as sample collection, quantification, quality control, and data interpretation. This review summarizes recent advances in targeted metabolomics and lipidomics, emphasizing their applications in clinical research. Advancements, including microsampling, dynamic multiple reaction monitoring, and integration of ion mobility mass spectrometry, are highlighted. Additionally, the review discusses the critical importance of data standardization and harmonization for successful clinical implementation.
Collapse
Affiliation(s)
- Nguyen Ky Anh
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam;
| | - Nguyen Quang Thu
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea (N.P.L.)
| | - Nguyen Tran Nam Tien
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea (N.P.L.)
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea (N.P.L.)
| | - Huy Truong Nguyen
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam;
| |
Collapse
|
29
|
Haue AD, Hjaltelin JX, Holm PC, Placido D, Brunak SR. Artificial intelligence-aided data mining of medical records for cancer detection and screening. Lancet Oncol 2024; 25:e694-e703. [PMID: 39637906 DOI: 10.1016/s1470-2045(24)00277-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 12/07/2024]
Abstract
The application of artificial intelligence methods to electronic patient records paves the way for large-scale analysis of multimodal data. Such population-wide data describing deep phenotypes composed of thousands of features are now being leveraged to create data-driven algorithms, which in turn has led to improved methods for early cancer detection and screening. Remaining challenges include establishment of infrastructures for prospective testing of such methods, ways to assess biases given the data, and gathering of sufficiently large and diverse datasets that reflect disease heterogeneities across populations. This Review provides an overview of artificial intelligence methods designed to detect cancer early, including key aspects of concern (eg, the problem of data drift-when the underlying health-care data change over time), ethical aspects, and discrepancies between access to cancer screening in high-income countries versus low-income and middle-income countries.
Collapse
Affiliation(s)
- Amalie Dahl Haue
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Copenhagen University Hospital Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jessica Xin Hjaltelin
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Christoffer Holm
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Davide Placido
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Copenhagen University Hospital Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - S Ren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Copenhagen University Hospital Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
30
|
Ren S, Li J, Dorado J, Sierra A, González-Díaz H, Duardo A, Shen B. From molecular mechanisms of prostate cancer to translational applications: based on multi-omics fusion analysis and intelligent medicine. Health Inf Sci Syst 2024; 12:6. [PMID: 38125666 PMCID: PMC10728428 DOI: 10.1007/s13755-023-00264-5] [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: 08/24/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Prostate cancer is the most common cancer in men worldwide and has a high mortality rate. The complex and heterogeneous development of prostate cancer has become a core obstacle in the treatment of prostate cancer. Simultaneously, the issues of overtreatment in early-stage diagnosis, oligometastasis and dormant tumor recognition, as well as personalized drug utilization, are also specific concerns that require attention in the clinical management of prostate cancer. Some typical genetic mutations have been proved to be associated with prostate cancer's initiation and progression. However, single-omic studies usually are not able to explain the causal relationship between molecular alterations and clinical phenotypes. Exploration from a systems genetics perspective is also lacking in this field, that is, the impact of gene network, the environmental factors, and even lifestyle behaviors on disease progression. At the meantime, current trend emphasizes the utilization of artificial intelligence (AI) and machine learning techniques to process extensive multidimensional data, including multi-omics. These technologies unveil the potential patterns, correlations, and insights related to diseases, thereby aiding the interpretable clinical decision making and applications, namely intelligent medicine. Therefore, there is a pressing need to integrate multidimensional data for identification of molecular subtypes, prediction of cancer progression and aggressiveness, along with perosonalized treatment performing. In this review, we systematically elaborated the landscape from molecular mechanism discovery of prostate cancer to clinical translational applications. We discussed the molecular profiles and clinical manifestations of prostate cancer heterogeneity, the identification of different states of prostate cancer, as well as corresponding precision medicine practices. Taking multi-omics fusion, systems genetics, and intelligence medicine as the main perspectives, the current research results and knowledge-driven research path of prostate cancer were summarized.
Collapse
Affiliation(s)
- Shumin Ren
- Department of Urology and Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041 China
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
| | - Jiakun Li
- Department of Urology and Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Julián Dorado
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
| | - Alejandro Sierra
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
- IKERDATA S.L., ZITEK, University of Basque Country UPVEHU, Rectorate Building, 48940 Leioa, Spain
| | - Humbert González-Díaz
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
- IKERDATA S.L., ZITEK, University of Basque Country UPVEHU, Rectorate Building, 48940 Leioa, Spain
| | - Aliuska Duardo
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
- IKERDATA S.L., ZITEK, University of Basque Country UPVEHU, Rectorate Building, 48940 Leioa, Spain
| | - Bairong Shen
- Department of Urology and Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041 China
| |
Collapse
|
31
|
Tan J, Xie J, Huang J, Deng W, Chai H, Yang Y. An interpretable survival model for diffuse large B-cell lymphoma patients using a biologically informed visible neural network. Comput Struct Biotechnol J 2024; 24:523-532. [PMID: 39211335 PMCID: PMC11357880 DOI: 10.1016/j.csbj.2024.07.019] [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: 01/19/2024] [Revised: 07/06/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin lymphoma (NHL) and is characterized by high heterogeneity. Assessment of its prognosis and genetic subtyping hold significant clinical implications. However, existing DLBCL prognostic models are mainly based on transcriptomic profiles, while genetic variation detection is more commonly used in clinical practice. In addition, current clustering-based subtyping methods mostly focus on genes with high mutation frequencies, providing insufficient explanations for the heterogeneity of DLBCL. Here, we proposed VNNSurv (https://bio-web1.nscc-gz.cn/app/VNNSurv), a survival model for DLBCL patients based on a biologically informed visible neural network (VNN). VNNSurv achieved an average C-index of 0.72 on the cross-validation set (HMRN cohort, n = 928), outperforming the baseline methods. The remarkable interpretability of VNNSurv facilitated the identification of the most impactful genes and the underlying pathways through which they act on patient outcomes. When only the 30 highest-impact genes were used as genetic input, the overall performance of VNNSurv improved, and a C-index of 0.70 was achieved on the external TCGA cohort (n = 48). Leveraging these high-impact genes, including 16 genes with low (<5 %) alteration frequencies, we devised a genetic-based prognostic index (GPI) for risk stratification and a subtype identification method. We stratified the patient group according to the International Prognostic Index (IPI) into three risk grades with significant prognostic differences. Furthermore, the defined subtypes exhibited greater prognostic consistency than clustering-based methods. Broadly, VNNSurv is a valuable DLBCL survival model. Its high interpretability has significant value for precision medicine, and its framework is scalable to other diseases.
Collapse
Affiliation(s)
- Jie Tan
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, China
- Guangzhou KingMed Center for Clinical Laboratory Co. Ltd., Guangzhou, China
| | - Jiancong Xie
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Jiarong Huang
- School of Mathematics and Big Data, Foshan University, Foshan, China
| | - Weizhen Deng
- School of Mathematics and Big Data, Foshan University, Foshan, China
| | - Hua Chai
- School of Mathematics and Big Data, Foshan University, Foshan, China
| | - Yuedong Yang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Machine Intelligence and Advanced Computing of MOE, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
32
|
Wang FA, Li Y, Zeng T. Deep Learning of radiology-genomics integration for computational oncology: A mini review. Comput Struct Biotechnol J 2024; 23:2708-2716. [PMID: 39035833 PMCID: PMC11260400 DOI: 10.1016/j.csbj.2024.06.019] [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: 03/06/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
In the field of computational oncology, patient status is often assessed using radiology-genomics, which includes two key technologies and data, such as radiology and genomics. Recent advances in deep learning have facilitated the integration of radiology-genomics data, and even new omics data, significantly improving the robustness and accuracy of clinical predictions. These factors are driving artificial intelligence (AI) closer to practical clinical applications. In particular, deep learning models are crucial in identifying new radiology-genomics biomarkers and therapeutic targets, supported by explainable AI (xAI) methods. This review focuses on recent developments in deep learning for radiology-genomics integration, highlights current challenges, and outlines some research directions for multimodal integration and biomarker discovery of radiology-genomics or radiology-omics that are urgently needed in computational oncology.
Collapse
Affiliation(s)
- Feng-ao Wang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yixue Li
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Guangzhou National Laboratory, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| | - Tao Zeng
- Guangzhou National Laboratory, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
33
|
Rai HM, Yoo J, Razaque A. Comparative analysis of machine learning and deep learning models for improved cancer detection: A comprehensive review of recent advancements in diagnostic techniques. EXPERT SYSTEMS WITH APPLICATIONS 2024; 255:124838. [DOI: 10.1016/j.eswa.2024.124838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
|
34
|
Cai Z, Poulos RC, Aref A, Robinson PJ, Reddel RR, Zhong Q. DeePathNet: A Transformer-Based Deep Learning Model Integrating Multiomic Data with Cancer Pathways. CANCER RESEARCH COMMUNICATIONS 2024; 4:3151-3164. [PMID: 39530738 PMCID: PMC11652962 DOI: 10.1158/2767-9764.crc-24-0285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 10/10/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
SIGNIFICANCE DeePathNet integrates cancer-specific biological pathways using transformer-based deep learning for enhanced cancer analysis. It outperforms existing models in predicting drug responses, cancer types, and subtypes. By enabling pathway-level biomarker discovery, DeePathNet represents a significant advancement in cancer research and could lead to more effective treatments.
Collapse
Affiliation(s)
- Zhaoxiang Cai
- ProCan, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Rebecca C. Poulos
- ProCan, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Adel Aref
- ProCan, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Phillip J. Robinson
- ProCan, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Roger R. Reddel
- ProCan, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Qing Zhong
- ProCan, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| |
Collapse
|
35
|
He F, Aebersold R, Baker MS, Bian X, Bo X, Chan DW, Chang C, Chen L, Chen X, Chen YJ, Cheng H, Collins BC, Corrales F, Cox J, E W, Van Eyk JE, Fan J, Faridi P, Figeys D, Gao GF, Gao W, Gao ZH, Goda K, Goh WWB, Gu D, Guo C, Guo T, He Y, Heck AJR, Hermjakob H, Hunter T, Iyer NG, Jiang Y, Jimenez CR, Joshi L, Kelleher NL, Li M, Li Y, Lin Q, Liu CH, Liu F, Liu GH, Liu Y, Liu Z, Low TY, Lu B, Mann M, Meng A, Moritz RL, Nice E, Ning G, Omenn GS, Overall CM, Palmisano G, Peng Y, Pineau C, Poon TCW, Purcell AW, Qiao J, Reddel RR, Robinson PJ, Roncada P, Sander C, Sha J, Song E, Srivastava S, Sun A, Sze SK, Tang C, Tang L, Tian R, Vizcaíno JA, Wang C, Wang C, Wang X, Wang X, Wang Y, Weiss T, Wilhelm M, Winkler R, Wollscheid B, Wong L, Xie L, Xie W, Xu T, Xu T, Yan L, Yang J, Yang X, Yates J, Yun T, Zhai Q, Zhang B, Zhang H, Zhang L, Zhang L, Zhang P, Zhang Y, Zheng YZ, Zhong Q, et alHe F, Aebersold R, Baker MS, Bian X, Bo X, Chan DW, Chang C, Chen L, Chen X, Chen YJ, Cheng H, Collins BC, Corrales F, Cox J, E W, Van Eyk JE, Fan J, Faridi P, Figeys D, Gao GF, Gao W, Gao ZH, Goda K, Goh WWB, Gu D, Guo C, Guo T, He Y, Heck AJR, Hermjakob H, Hunter T, Iyer NG, Jiang Y, Jimenez CR, Joshi L, Kelleher NL, Li M, Li Y, Lin Q, Liu CH, Liu F, Liu GH, Liu Y, Liu Z, Low TY, Lu B, Mann M, Meng A, Moritz RL, Nice E, Ning G, Omenn GS, Overall CM, Palmisano G, Peng Y, Pineau C, Poon TCW, Purcell AW, Qiao J, Reddel RR, Robinson PJ, Roncada P, Sander C, Sha J, Song E, Srivastava S, Sun A, Sze SK, Tang C, Tang L, Tian R, Vizcaíno JA, Wang C, Wang C, Wang X, Wang X, Wang Y, Weiss T, Wilhelm M, Winkler R, Wollscheid B, Wong L, Xie L, Xie W, Xu T, Xu T, Yan L, Yang J, Yang X, Yates J, Yun T, Zhai Q, Zhang B, Zhang H, Zhang L, Zhang L, Zhang P, Zhang Y, Zheng YZ, Zhong Q, Zhu Y. π-HuB: the proteomic navigator of the human body. Nature 2024; 636:322-331. [PMID: 39663494 DOI: 10.1038/s41586-024-08280-5] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 10/23/2024] [Indexed: 12/13/2024]
Abstract
The human body contains trillions of cells, classified into specific cell types, with diverse morphologies and functions. In addition, cells of the same type can assume different states within an individual's body during their lifetime. Understanding the complexities of the proteome in the context of a human organism and its many potential states is a necessary requirement to understanding human biology, but these complexities can neither be predicted from the genome, nor have they been systematically measurable with available technologies. Recent advances in proteomic technology and computational sciences now provide opportunities to investigate the intricate biology of the human body at unprecedented resolution and scale. Here we introduce a big-science endeavour called π-HuB (proteomic navigator of the human body). The aim of the π-HuB project is to (1) generate and harness multimodality proteomic datasets to enhance our understanding of human biology; (2) facilitate disease risk assessment and diagnosis; (3) uncover new drug targets; (4) optimize appropriate therapeutic strategies; and (5) enable intelligent healthcare, thereby ushering in a new era of proteomics-driven phronesis medicine. This ambitious mission will be implemented by an international collaborative force of multidisciplinary research teams worldwide across academic, industrial and government sectors.
Collapse
Affiliation(s)
- Fuchu He
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
- International Academy of Phronesis Medicine (Guangdong), Guangdong, China.
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
| | - Mark S Baker
- Macquarie Medical School, Macquarie University, Sydney, New South Wales, Australia
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xiaochen Bo
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Daniel W Chan
- Department of Pathology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Cheng Chang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Yu-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei, China
| | - Heping Cheng
- National Biomedical Imaging Center, State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, College of Future Technology, Peking University, Beijing, China
| | - Ben C Collins
- School of Biological Sciences, Queen's University of Belfast, Belfast, UK
| | - Fernando Corrales
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología-CSIC, Madrid, Spain
| | - Jürgen Cox
- Computational Systems Biochemistry Research Group, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Weinan E
- AI for Science Institute, Beijing, China
- Center for Machine Learning Research, Peking University, Beijing, China
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Pouya Faridi
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Monash Proteomics and Metabolomics Platform, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Daniel Figeys
- School of Pharmaceutical Sciences and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - George Fu Gao
- The D. H. Chen School of Universal Health, Zhejiang University, Hangzhou, China
| | - Wen Gao
- Pengcheng Laboratory, Shenzhen, China
- School of Electronic Engineering and Computer Science, Peking University, Beijing, China
| | - Zu-Hua Gao
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Keisuke Goda
- Department of Chemistry, University of Tokyo, Tokyo, Japan
- Department of Bioengineering, University of California, Los Angeles, California, USA
- Institute of Technological Sciences, Wuhan University, Wuhan, Hubei, China
| | - Wilson Wen Bin Goh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Dongfeng Gu
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Changjiang Guo
- Department of Nutrition, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Tiannan Guo
- School of Medicine, Westlake University, Hangzhou, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China
| | - Yuezhong He
- International Academy of Phronesis Medicine (Guangdong), Guangdong, China
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, the Netherlands
- Netherlands Proteomics Center, Utrecht, the Netherlands
| | - Henning Hermjakob
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Narayanan Gopalakrishna Iyer
- Department of Head & Neck Surgery, Division of Surgery & Surgical Oncology, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Ying Jiang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Connie R Jimenez
- OncoProteomics Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Lokesh Joshi
- Advanced Glycoscience Research Cluster, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Neil L Kelleher
- Departments of Molecular Biosciences, Departments of Chemistry, Northwestern University, Evanston, IL, USA
| | - Ming Li
- David R. Cheriton School of Computer Science, University of Waterloo, Waterloo, Ontario, Canada
- Central China Institute of Artificial Intelligence, Henan, China
| | - Yang Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Qingsong Lin
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Fan Liu
- Department of Structural Biology, Leibniz-Forschungsinstitut für MolekularePharmakologie (FMP), Berlin, Germany
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yansheng Liu
- Cancer Biology Institute, Yale University School of Medicine, West Haven, CT, USA
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ben Lu
- Department of Critical Care Medicine and Hematology, The Third Xiangya Hospital, Central South University; Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Matthias Mann
- Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Anming Meng
- School of Life Sciences, Tsinghua University, Tsinghua-Peking Center for Life Sciences, Beijing, China
| | | | - Edouard Nice
- Clinical Biomarker Discovery and Validation, Monash University, Clayton, Victoria, Australia
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gilbert S Omenn
- Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Christopher M Overall
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Yonsei Frontier Lab, Yonsei University, Seoul, Republic of Korea
| | - Giuseppe Palmisano
- Glycoproteomics Laboratory, Department of Parasitology, University of São Paulo, Sao Paulo, Brazil
| | - Yaojin Peng
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Charles Pineau
- Institut de Recherche en Santé Environnement et Travail, Univ. Rennes, Inserm, EHESP, Irset, Rennes, France
| | - Terence Chuen Wai Poon
- Pilot Laboratory, MOE Frontier Science Centre for Precision Oncology, Centre for Precision Medicine Research and Training, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Anthony W Purcell
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Roger R Reddel
- ProCan®, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
| | - Phillip J Robinson
- ProCan®, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
| | - Paola Roncada
- Department of Health Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Chris Sander
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jiahao Sha
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | | | - Aihua Sun
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Siu Kwan Sze
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Chao Tang
- Center for Quantitative Biology, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Liujun Tang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Ruijun Tian
- Department of Chemistry, Southern University of Science and Technology, Shenzhen, China
| | - Juan Antonio Vizcaíno
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Chanjuan Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Chen Wang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
| | - Xiaowen Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Xinxing Wang
- Department of Nutrition, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Yan Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Tobias Weiss
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | | | - Robert Winkler
- Advanced Genomics Unit, Center for Research and Advanced Studies, Irapuato, Mexico
| | - Bernd Wollscheid
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Limsoon Wong
- Department of Computer Science, National University of Singapore, Singapore, Singapore
- Department of Pathology, National University of Singapore, Singapore, Singapore
| | - Linhai Xie
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Wei Xie
- School of Life Sciences, Tsinghua University, Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Tao Xu
- Guangzhou National Laboratory, Guangzhou, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, China
| | - Tianhao Xu
- International Academy of Phronesis Medicine (Guangdong), Guangdong, China
| | - Liying Yan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Jing Yang
- Guangzhou National Laboratory, Guangzhou, China
| | - Xiao Yang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - John Yates
- The Scripps Research Institute, La Jolla, CA, USA
| | - Tao Yun
- China Science and Technology Exchange Center, Beijing, China
| | - Qiwei Zhai
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Hui Zhang
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Lihua Zhang
- State Key Laboratory of Medical Proteomics, National Chromatography R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Lingqiang Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Pingwen Zhang
- School of Mathematical Sciences, Peking University, Beijing, China
- Wuhan University, Wuhan, China
| | - Yukui Zhang
- State Key Laboratory of Medical Proteomics, National Chromatography R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yu Zi Zheng
- International Academy of Phronesis Medicine (Guangdong), Guangdong, China
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Qing Zhong
- ProCan®, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
| | - Yunping Zhu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
36
|
Niarakis A, Laubenbacher R, An G, Ilan Y, Fisher J, Flobak Å, Reiche K, Rodríguez Martínez M, Geris L, Ladeira L, Veschini L, Blinov ML, Messina F, Fonseca LL, Ferreira S, Montagud A, Noël V, Marku M, Tsirvouli E, Torres MM, Harris LA, Sego TJ, Cockrell C, Shick AE, Balci H, Salazar A, Rian K, Hemedan AA, Esteban-Medina M, Staumont B, Hernandez-Vargas E, Martis B S, Madrid-Valiente A, Karampelesis P, Sordo Vieira L, Harlapur P, Kulesza A, Nikaein N, Garira W, Malik Sheriff RS, Thakar J, Tran VDT, Carbonell-Caballero J, Safaei S, Valencia A, Zinovyev A, Glazier JA. Immune digital twins for complex human pathologies: applications, limitations, and challenges. NPJ Syst Biol Appl 2024; 10:141. [PMID: 39616158 PMCID: PMC11608242 DOI: 10.1038/s41540-024-00450-5] [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: 05/27/2024] [Accepted: 09/27/2024] [Indexed: 12/06/2024] Open
Abstract
Digital twins represent a key technology for precision health. Medical digital twins consist of computational models that represent the health state of individual patients over time, enabling optimal therapeutics and forecasting patient prognosis. Many health conditions involve the immune system, so it is crucial to include its key features when designing medical digital twins. The immune response is complex and varies across diseases and patients, and its modelling requires the collective expertise of the clinical, immunology, and computational modelling communities. This review outlines the initial progress on immune digital twins and the various initiatives to facilitate communication between interdisciplinary communities. We also outline the crucial aspects of an immune digital twin design and the prerequisites for its implementation in the clinic. We propose some initial use cases that could serve as "proof of concept" regarding the utility of immune digital technology, focusing on diseases with a very different immune response across spatial and temporal scales (minutes, days, months, years). Lastly, we discuss the use of digital twins in drug discovery and point out emerging challenges that the scientific community needs to collectively overcome to make immune digital twins a reality.
Collapse
Affiliation(s)
- Anna Niarakis
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, UPS, CNRS, Toulouse, France.
- Lifeware Group, Inria, Saclay-île de France, Palaiseau, France.
| | | | - Gary An
- Department of Surgery, University of Vermont Larner College of Medicine, Vermont, USA
| | - Yaron Ilan
- Faculty of Medicine Hebrew University, Hadassah Medical Center, Jerusalem, Israel
| | - Jasmin Fisher
- UCL Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6BT, UK
| | - Åsmund Flobak
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- The Cancer Clinic, St Olav's University Hospital, Trondheim, Norway
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Kristin Reiche
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- Institute of Clinical Immunology, Medical Faculty, University Hospital, University of Leipzig, Leipzig, Germany
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), Dresden/Leipzig, Germany
| | - María Rodríguez Martínez
- Department of Biomedical Informatics & Data Science, Yale School of Medicine, New Haven, CT, USA
| | - Liesbet Geris
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Biomechanics Research Unit, GIGA Molecular and Computational Biology, University of Liège, Liège, Belgium
| | - Luiz Ladeira
- Biomechanics Research Unit, GIGA Molecular and Computational Biology, University of Liège, Liège, Belgium
| | - Lorenzo Veschini
- Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, London, UK
- Biocomplexity Institute and Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, 47408, USA
| | - Michael L Blinov
- Center for Cell Analysis and Modeling, UConn Health, Farmington, CT, 06030, USA
| | - Francesco Messina
- Department of Epidemiology, Preclinical Research and Advanced Diagnostic, National Institute for Infectious Diseases 'Lazzaro Spallanzani' - I.R.C.C.S., Rome, Italy
| | - Luis L Fonseca
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Sandra Ferreira
- Mathematics Department and Center of Mathematics, University of Beira Interior, Covilhã, Portugal
| | - Arnau Montagud
- Barcelona Supercomputing Center (BSC), Barcelone, Spain
- Institute for Integrative Systems Biology (I2SysBio), CSIC-UV, Valencia, Spain
| | - Vincent Noël
- Institut Curie, Université PSL, F-75005, Paris, France
- INSERM, U900, F-75005, Paris, France
- Mines ParisTech, Université PSL, F-75005, Paris, France
| | - Malvina Marku
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Eirini Tsirvouli
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marcella M Torres
- Department of Mathematics and Statistics, University of Richmond, Richmond, VA, USA
| | - Leonard A Harris
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
- Interdisciplinary Graduate Program in Cell and Molecular Biology, University of Arkansas, Fayetteville, AR, USA
- Cancer Biology Program, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - T J Sego
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Chase Cockrell
- Department of Surgery, University of Vermont Larner College of Medicine, Vermont, USA
| | - Amanda E Shick
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL, USA
| | - Hasan Balci
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, The Netherlands
| | - Albin Salazar
- INRIA Paris/CNRS/École Normale Supérieure/PSL Research University, Paris, France
| | - Kinza Rian
- Andalusian Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
| | - Ahmed Abdelmonem Hemedan
- Bioinformatics Core Unit, Luxembourg Centre of Systems Biomedicine LCSB, Luxembourg University, Esch-sur-Alzette, Luxembourg
| | - Marina Esteban-Medina
- Andalusian Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
| | - Bernard Staumont
- Biomechanics Research Unit, GIGA Molecular and Computational Biology, University of Liège, Liège, Belgium
| | - Esteban Hernandez-Vargas
- Department of Mathematics and Statistical Science, University of Idaho, Moscow, ID, 83844-1103, USA
| | | | | | | | | | - Pradyumna Harlapur
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India
| | | | - Niloofar Nikaein
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, SE-70182, Örebro, Sweden
- X-HiDE - Exploring Inflammation in Health and Disease Consortium, Örebro University, Örebro, Sweden
| | - Winston Garira
- Multiscale Mathematical Modelling of Living Systems program (M3-LSP), Kimberley, South Africa
- Department of Mathematical Sciences, Sol Plaatje University, Kimberley, South Africa
- Private Bag X5008, Kimberley, 8300, South Africa
| | - Rahuman S Malik Sheriff
- European Bioinformatics Institute, European Molecular Biology Laboratory (EMBL-EBI), Hinxton, Cambridge, UK
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Juilee Thakar
- Department of Microbiology & Immunology and Department of Biostatistics & Computational Biology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Van Du T Tran
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Soroush Safaei
- Institute of Biomedical Engineering and Technology, Ghent University, Gent, Belgium
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC), Barcelone, Spain
- ICREA, 23 Passeig Lluís Companys, 08010, Barcelona, Spain
| | | | - James A Glazier
- Biocomplexity Institute and Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, 47408, USA
| |
Collapse
|
37
|
Oldenburg J, Wagner J, Troschke-Meurer S, Plietz J, Kaderali L, Völzke H, Nauck M, Homuth G, Völker U, Simm S. XModNN: Explainable Modular Neural Network to Identify Clinical Parameters and Disease Biomarkers in Transcriptomic Datasets. Biomolecules 2024; 14:1501. [PMID: 39766207 PMCID: PMC11673716 DOI: 10.3390/biom14121501] [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: 10/24/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
The Explainable Modular Neural Network (XModNN) enables the identification of biomarkers, facilitating the classification of diseases and clinical parameters in transcriptomic datasets. The modules within XModNN represent specific pathways or genes of a functional hierarchy. The incorporation of biological insights into the architectural design reduced the number of parameters. This is further reinforced by the weighted multi-loss progressive training, which enables successful classification with a reduced number of replicates. The combination of this workflow with layer-wise relevance propagation ensures a robust post hoc explanation of the individual module contribution. Two use cases were employed to predict sex and neuroblastoma cell states, demonstrating that XModNN, in contrast to standard statistical approaches, results in a reduced number of candidate biomarkers. Moreover, the architecture enables the training on a limited number of examples, attaining the same performance and robustness as support vector machine and random forests. The integrated pathway relevance analysis improves a standard gene set overrepresentation analysis, which relies solely on gene assignment. Two crucial genes and three pathways were identified for sex classification, while 26 genes and six pathways are highly important to discriminate adrenergic-mesenchymal cell states in neuroblastoma cancer.
Collapse
Affiliation(s)
- Jan Oldenburg
- Institute of Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany; (J.O.); (J.W.); (L.K.)
- Institute for Bioanalysis, Department of Applied Sciences, Coburg University of Applied Sciences and Arts, 96450 Coburg, Germany
| | - Jonas Wagner
- Institute of Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany; (J.O.); (J.W.); (L.K.)
| | - Sascha Troschke-Meurer
- Institute for Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.T.-M.); (J.P.)
| | - Jessica Plietz
- Institute for Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.T.-M.); (J.P.)
| | - Lars Kaderali
- Institute of Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany; (J.O.); (J.W.); (L.K.)
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany; (G.H.); (U.V.)
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany; (G.H.); (U.V.)
| | - Stefan Simm
- Institute of Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany; (J.O.); (J.W.); (L.K.)
- Institute for Bioanalysis, Department of Applied Sciences, Coburg University of Applied Sciences and Arts, 96450 Coburg, Germany
| |
Collapse
|
38
|
Hsieh KL, Zhang K, Chu Y, Yu L, Li X, Hu N, Kawosa I, Pilié PG, Bhattacharya PK, Zhi D, Jiang X, Zhao Z, Dai Y. iGTP: Learning interpretable cellular embedding for inferring biological mechanisms underlying single-cell transcriptomics. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.29.24305092. [PMID: 39649598 PMCID: PMC11623718 DOI: 10.1101/2024.03.29.24305092] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Deep-learning models like Variational AutoEncoder have enabled low dimensional cellular embedding representation for large-scale single-cell transcriptomes and shown great flexibility in downstream tasks. However, biologically meaningful latent space is usually missing if no specific structure is designed. Here, we engineered a novel interpretable generative transcriptional program (iGTP) framework that could model the importance of transcriptional program (TP) space and protein-protein interactions (PPI) between different biological states. We demonstrated the performance of iGTP in a diverse biological context using gene ontology, canonical pathway, and different PPI curation. iGTP not only elucidated the ground truth of cellular responses but also surpassed other deep learning models and traditional bioinformatics methods in functional enrichment tasks. By integrating the latent layer with a graph neural network framework, iGTP could effectively infer cellular responses to perturbations. Lastly, we applied iGTP TP embeddings with a latent diffusion model to accurately generate cell embeddings for specific cell types and states. We anticipate that iGTP will offer insights at both PPI and TP levels and holds promise for predicting responses to novel perturbations.
Collapse
|
39
|
Wu Y, Xu P, Wang L, Liu S, Hou Y, Lu H, Hu P, Li X, Yu X. scGO: interpretable deep neural network for cell status annotation and disease diagnosis. Brief Bioinform 2024; 26:bbaf018. [PMID: 39820437 PMCID: PMC11737892 DOI: 10.1093/bib/bbaf018] [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: 09/02/2024] [Revised: 12/16/2024] [Accepted: 01/10/2025] [Indexed: 01/19/2025] Open
Abstract
Machine learning has emerged as a transformative tool for elucidating cellular heterogeneity in single-cell RNA sequencing. However, a significant challenge lies in the "black box" nature of deep learning models, which obscures the decision-making process and limits interpretability in cell status annotation. In this study, we introduced scGO, a Gene Ontology (GO)-inspired deep learning framework designed to provide interpretable cell status annotation for scRNA-seq data. scGO employs sparse neural networks to leverage the intrinsic biological relationships among genes, transcription factors, and GO terms, significantly augmenting interpretability and reducing computational cost. scGO outperforms state-of-the-art methods in the precise characterization of cell subtypes across diverse datasets. Our extensive experimentation across a spectrum of scRNA-seq datasets underscored the remarkable efficacy of scGO in disease diagnosis, prediction of developmental stages, and evaluation of disease severity and cellular senescence status. Furthermore, we incorporated in silico individual gene manipulations into the scGO model, introducing an additional layer for discovering therapeutic targets. Our results provide an interpretable model for accurately annotating cell status, capturing latent biological knowledge, and informing clinical practice.
Collapse
Affiliation(s)
- You Wu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dong Chuan Road, Shanghai 200240, China
| | - Pengfei Xu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dong Chuan Road, Shanghai 200240, China
| | - Liyuan Wang
- School of Agriculture and Biology, Shanghai Jiao Tong University, No. 800 Dong Chuan Road, Shanghai 200240, China
| | - Shuai Liu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dong Chuan Road, Shanghai 200240, China
| | - Yingnan Hou
- School of Agriculture and Biology, Shanghai Jiao Tong University, No. 800 Dong Chuan Road, Shanghai 200240, China
| | - Hui Lu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dong Chuan Road, Shanghai 200240, China
| | - Peng Hu
- Ministry of Education, Shanghai Ocean University, No. 999, Huchenghuan Road, Shanghai 201306, China
| | - Xiaofei Li
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dong Chuan Road, Shanghai 200240, China
- Shanghai Pudong New Area People’s Hospital, No. 490, Chuanhuan South Road, Shanghai 201299, China
| | - Xiang Yu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, No. 800 Dong Chuan Road, Shanghai 200240, China
| |
Collapse
|
40
|
Jemimah S, Abuhantash F, AlShehhi A. c-Triadem: A constrained, explainable deep learning model to identify novel biomarkers in Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.19.24317595. [PMID: 39606415 PMCID: PMC11601769 DOI: 10.1101/2024.11.19.24317595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that requires early diagnosis for effective management. However, issues with currently available diagnostic biomarkers preclude early diagnosis, necessitating the development of alternative biomarkers and methods, such as blood-based diagnostics. We propose c-Triadem (constrained triple-input Alzheimer's disease model), a novel deep neural network to identify potential blood-based biomarkers for AD and predict mild cognitive impairment (MCI) and AD with high accuracy. The model utilizes genotyping data, gene expression data, and clinical information to predict the disease status of participants, i.e., cognitively normal (CN), MCI, or AD. The nodes of the neural network represent genes and their related pathways, and the edges represent known relationships among the genes and pathways. We trained the model with blood genotyping data, microarray, and clinical features from the Alzheimer's Neuroimaging Disease Initiative (ADNI). We demonstrate that our model's performance is superior to previous models with an AUC of 97% and accuracy of 89%. We then identified the most influential genes and clinical features for prediction using SHapley Additive exPlanations (SHAP). Our SHAP analysis shows that CASP9, LCK, and SDC3 SNPs and PINK1, ATG5, and ubiquitin (UBB, UBC) expression have a higher impact on model performance. Our model has facilitated the identification of potential blood-based genetic markers of DNA damage response and mitophagy in affected regions of the brain. The model can be used for detection and biomarker identification in other related dementias.
Collapse
Affiliation(s)
- Sherlyn Jemimah
- Department of Biomedical Engineering and Biotechnology, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates
| | - Ferial Abuhantash
- Department of Biomedical Engineering and Biotechnology, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates
| | - Aamna AlShehhi
- Department of Biomedical Engineering and Biotechnology, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates
- Healthcare Engineering Innovation Center, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates
| |
Collapse
|
41
|
Zhong S, Su L, Xu M, Loke D, Yu B, Zhang Y, Zhao R. Recent Advances in Artificial Sensory Neurons: Biological Fundamentals, Devices, Applications, and Challenges. NANO-MICRO LETTERS 2024; 17:61. [PMID: 39537845 PMCID: PMC11561216 DOI: 10.1007/s40820-024-01550-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/28/2024] [Indexed: 11/16/2024]
Abstract
Spike-based neural networks, which use spikes or action potentials to represent information, have gained a lot of attention because of their high energy efficiency and low power consumption. To fully leverage its advantages, converting the external analog signals to spikes is an essential prerequisite. Conventional approaches including analog-to-digital converters or ring oscillators, and sensors suffer from high power and area costs. Recent efforts are devoted to constructing artificial sensory neurons based on emerging devices inspired by the biological sensory system. They can simultaneously perform sensing and spike conversion, overcoming the deficiencies of traditional sensory systems. This review summarizes and benchmarks the recent progress of artificial sensory neurons. It starts with the presentation of various mechanisms of biological signal transduction, followed by the systematic introduction of the emerging devices employed for artificial sensory neurons. Furthermore, the implementations with different perceptual capabilities are briefly outlined and the key metrics and potential applications are also provided. Finally, we highlight the challenges and perspectives for the future development of artificial sensory neurons.
Collapse
Affiliation(s)
- Shuai Zhong
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, 519031, People's Republic of China.
| | - Lirou Su
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, 519031, People's Republic of China
| | - Mingkun Xu
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, 519031, People's Republic of China
| | - Desmond Loke
- Department of Science, Mathematics and Technology, Singapore University of Technology and Design, Singapore, 487372, Singapore
| | - Bin Yu
- College of Integrated Circuits, Zhejiang University, Hangzhou, 3112000, People's Republic of China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 310027, People's Republic of China
| | - Yishu Zhang
- College of Integrated Circuits, Zhejiang University, Hangzhou, 3112000, People's Republic of China.
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 310027, People's Republic of China.
| | - Rong Zhao
- Department of Precision Instruments, Tsinghua University, Beijing, 100084, People's Republic of China
- Center for Brain-Inspired Computing Research, Tsinghua University, Beijing, 100084, People's Republic of China
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, People's Republic of China
| |
Collapse
|
42
|
Wang J, Wen Y, Zhang Y, Wang Z, Jiang Y, Dai C, Wu L, Leng D, He S, Bo X. An interpretable artificial intelligence framework for designing synthetic lethality-based anti-cancer combination therapies. J Adv Res 2024; 65:329-343. [PMID: 38043609 PMCID: PMC11519055 DOI: 10.1016/j.jare.2023.11.035] [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: 09/05/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/05/2023] Open
Abstract
INTRODUCTION Synthetic lethality (SL) provides an opportunity to leverage different genetic interactions when designing synergistic combination therapies. To further explore SL-based combination therapies for cancer treatment, it is important to identify and mechanistically characterize more SL interactions. Artificial intelligence (AI) methods have recently been proposed for SL prediction, but the results of these models are often not interpretable such that deriving the underlying mechanism can be challenging. OBJECTIVES This study aims to develop an interpretable AI framework for SL prediction and subsequently utilize it to design SL-based synergistic combination therapies. METHODS We propose a knowledge and data dual-driven AI framework for SL prediction (KDDSL). Specifically, we use gene knowledge related to the SL mechanism to guide the construction of the model and develop a method to identify the most relevant gene knowledge for the predicted results. RESULTS Experimental and literature-based validation confirmed a good balance between predictive and interpretable ability when using KDDSL. Moreover, we demonstrated that KDDSL could help to discover promising drug combinations and clarify associated biological processes, such as the combination of MDM2 and CDK9 inhibitors, which exhibited significant anti-cancer effects in vitro and in vivo. CONCLUSION These data underscore the potential of KDDSL to guide SL-based combination therapy design. There is a need for biomedicine-focused AI strategies to combine rational biological knowledge with developed models.
Collapse
Affiliation(s)
- Jing Wang
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yuqi Wen
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Yixin Zhang
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Zhongming Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Yuyang Jiang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Chong Dai
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Lianlian Wu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Dongjin Leng
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Song He
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.
| | - Xiaochen Bo
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.
| |
Collapse
|
43
|
Wu Q, Morrow EM, Gamsiz Uzun ED. A deep learning model for prediction of autism status using whole-exome sequencing data. PLoS Comput Biol 2024; 20:e1012468. [PMID: 39514604 DOI: 10.1371/journal.pcbi.1012468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 11/20/2024] [Accepted: 09/06/2024] [Indexed: 11/16/2024] Open
Abstract
Autism is a developmental disability. Research demonstrated that children with autism benefit from early diagnosis and early intervention. Genetic factors are considered major contributors to the development of autism. Machine learning (ML), including deep learning (DL), has been evaluated in phenotype prediction, but this method has been limited in its application to autism. We developed a DL model, the Separate Translated Autism Research Neural Network (STAR-NN) model to predict autism status. The model was trained and tested using whole exome sequencing data from 43,203 individuals (16,809 individuals with autism and 26,394 non-autistic controls). Polygenic scores from common variants and the aggregated count of rare variants on genes were used as input. In STAR-NN, protein truncating variants, possibly damaging missense variants and mild effect missense variants on the same gene were separated at the input level and merged to one gene node. In this way, rare variants with different level of pathogenic effects were treated separately. We further validated the performance of STAR-NN using an independent dataset, including 13,827 individuals with autism and 14,052 non-autistic controls. STAR-NN achieved a modest ROC-AUC of 0.7319 on the testing dataset and 0.7302 on the independent dataset. STAR-NN outperformed other traditional ML models. Gene Ontology analysis on the selected gene features showed an enrichment for potentially informative pathways including calcium ion transport.
Collapse
Affiliation(s)
- Qing Wu
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, United States of America
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| | - Eric M Morrow
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, United States of America
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
- Developmental Disorders Genetics Research Program, Department of Psychiatry and Human Behavior, Emma Pendleton Bradley Hospital, East Providence, Rhode Island, United States of America
| | - Ece D Gamsiz Uzun
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, Rhode Island, United States of America
| |
Collapse
|
44
|
Bao R, Hutson A, Madabhushi A, Jonsson VD, Rosario SR, Barnholtz-Sloan JS, Fertig EJ, Marathe H, Harris L, Altreuter J, Chen Q, Dignam J, Gentles AJ, Gonzalez-Kozlova E, Gnjatic S, Kim E, Long M, Morgan M, Ruppin E, Valen DV, Zhang H, Vokes N, Meerzaman D, Liu S, Van Allen EM, Xing Y. Ten challenges and opportunities in computational immuno-oncology. J Immunother Cancer 2024; 12:e009721. [PMID: 39461879 PMCID: PMC11529678 DOI: 10.1136/jitc-2024-009721] [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: 06/05/2024] [Accepted: 09/23/2024] [Indexed: 10/29/2024] Open
Abstract
Immuno-oncology has transformed the treatment of cancer, with several immunotherapies becoming the standard treatment across histologies. Despite these advancements, the majority of patients do not experience durable clinical benefits, highlighting the imperative for ongoing advancement in immuno-oncology. Computational immuno-oncology emerges as a forefront discipline that draws on biomedical data science and intersects with oncology, immunology, and clinical research, with the overarching goal to accelerate the development of effective and safe immuno-oncology treatments from the laboratory to the clinic. In this review, we outline 10 critical challenges and opportunities in computational immuno-oncology, emphasizing the importance of robust computational strategies and interdisciplinary collaborations amid the constantly evolving interplay between clinical needs and technological innovation.
Collapse
Affiliation(s)
- Riyue Bao
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alan Hutson
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Anant Madabhushi
- Emory University, Atlanta, Georgia, USA
- Georgia Institute of Technology, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Medical Center, Atlanta, Georgia, USA
| | - Vanessa D Jonsson
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, California, USA
| | - Spencer R Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Jill S Barnholtz-Sloan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, Bethesda, Maryland, USA
| | - Elana J Fertig
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Himangi Marathe
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Lyndsay Harris
- Cancer Diagnosis Program, National Cancer Institute Division of Cancer Treatment and Diagnosis, Bethesda, Maryland, USA
| | | | - Qingrong Chen
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, Maryland, USA
| | - James Dignam
- Department of Public Health Sciences, University of Chicago Division of the Biological Sciences, Chicago, Illinois, USA
| | - Andrew J Gentles
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Edgar Gonzalez-Kozlova
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Immunology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sacha Gnjatic
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Immunology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Erika Kim
- Informatics and Data Science Program, Center for Biomedical Informatics and Information Technology, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Martin Morgan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, Maryland, USA
| | - David Van Valen
- Division of Computing and Mathematical Science, Caltech, Pasadena, California, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Hong Zhang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Natalie Vokes
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daoud Meerzaman
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, Maryland, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Eliezer M Van Allen
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yi Xing
- Center for Computational and Genomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
45
|
Wang H, Argenziano MG, Yoon H, Boyett D, Save A, Petridis P, Savage W, Jackson P, Hawkins-Daarud A, Tran N, Hu L, Singleton KW, Paulson L, Dalahmah OA, Bruce JN, Grinband J, Swanson KR, Canoll P, Li J. Biologically informed deep neural networks provide quantitative assessment of intratumoral heterogeneity in post treatment glioblastoma. NPJ Digit Med 2024; 7:292. [PMID: 39427044 PMCID: PMC11490546 DOI: 10.1038/s41746-024-01277-4] [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: 03/06/2024] [Accepted: 09/27/2024] [Indexed: 10/21/2024] Open
Abstract
Intratumoral heterogeneity poses a significant challenge to the diagnosis and treatment of recurrent glioblastoma. This study addresses the need for non-invasive approaches to map heterogeneous landscape of histopathological alterations throughout the entire lesion for each patient. We developed BioNet, a biologically-informed neural network, to predict regional distributions of two primary tissue-specific gene modules: proliferating tumor (Pro) and reactive/inflammatory cells (Inf). BioNet significantly outperforms existing methods (p < 2e-26). In cross-validation, BioNet achieved AUCs of 0.80 (Pro) and 0.81 (Inf), with accuracies of 80% and 75%, respectively. In blind tests, BioNet achieved AUCs of 0.80 (Pro) and 0.76 (Inf), with accuracies of 81% and 74%. Competing methods had AUCs lower or around 0.6 and accuracies lower or around 70%. BioNet's voxel-level prediction maps reveal intratumoral heterogeneity, potentially improving biopsy targeting and treatment evaluation. This non-invasive approach facilitates regular monitoring and timely therapeutic adjustments, highlighting the role of ML in precision medicine.
Collapse
Affiliation(s)
- Hairong Wang
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael G Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Hyunsoo Yoon
- Department of Industrial Engineering, Yonsei University, Seoul, South Korea
| | - Deborah Boyett
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Akshay Save
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Petros Petridis
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Psychiatry, New York University, New York, NY, USA
| | - William Savage
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Pamela Jackson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - Andrea Hawkins-Daarud
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - Nhan Tran
- Department of Cancer Biology, Mayo Clinic, Phoenix, AZ, USA
| | - Leland Hu
- Department of Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Kyle W Singleton
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - Lisa Paulson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - Osama Al Dalahmah
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Jack Grinband
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kristin R Swanson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jing Li
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
46
|
Sun X, Guo X, Gao J, Wu J, Huang F, Zhang JH, Huang F, Lu X, Shi Y, Pan L. E-Skin and Its Advanced Applications in Ubiquitous Health Monitoring. Biomedicines 2024; 12:2307. [PMID: 39457619 PMCID: PMC11505155 DOI: 10.3390/biomedicines12102307] [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: 08/30/2024] [Revised: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
E-skin is a bionic device with flexible and intelligent sensing ability that can mimic the touch, temperature, pressure, and other sensing functions of human skin. Because of its flexibility, breathability, biocompatibility, and other characteristics, it is widely used in health management, personalized medicine, disease prevention, and other pan-health fields. With the proposal of new sensing principles, the development of advanced functional materials, the development of microfabrication technology, and the integration of artificial intelligence and algorithms, e-skin has developed rapidly. This paper focuses on the characteristics, fundamentals, new principles, key technologies, and their specific applications in health management, exercise monitoring, emotion and heart monitoring, etc. that advanced e-skin needs to have in the healthcare field. In addition, its significance in infant and child care, elderly care, and assistive devices for the disabled is analyzed. Finally, the current challenges and future directions of the field are discussed. It is expected that this review will generate great interest and inspiration for the development and improvement of novel e-skins and advanced health monitoring systems.
Collapse
Affiliation(s)
- Xidi Sun
- Collaborative Innovation Center of Advanced Microstructures, School of Electronic Science and Engineering, Nanjing University, Nanjing 210093, China; (X.S.); (X.G.); (J.G.); (J.W.); (F.H.)
| | - Xin Guo
- Collaborative Innovation Center of Advanced Microstructures, School of Electronic Science and Engineering, Nanjing University, Nanjing 210093, China; (X.S.); (X.G.); (J.G.); (J.W.); (F.H.)
| | - Jiansong Gao
- Collaborative Innovation Center of Advanced Microstructures, School of Electronic Science and Engineering, Nanjing University, Nanjing 210093, China; (X.S.); (X.G.); (J.G.); (J.W.); (F.H.)
| | - Jing Wu
- Collaborative Innovation Center of Advanced Microstructures, School of Electronic Science and Engineering, Nanjing University, Nanjing 210093, China; (X.S.); (X.G.); (J.G.); (J.W.); (F.H.)
| | - Fengchang Huang
- Collaborative Innovation Center of Advanced Microstructures, School of Electronic Science and Engineering, Nanjing University, Nanjing 210093, China; (X.S.); (X.G.); (J.G.); (J.W.); (F.H.)
| | - Jia-Han Zhang
- School of Electronic Information Engineering, Inner Mongolia University, Hohhot 010021, China;
| | - Fuhua Huang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China;
| | - Xiao Lu
- The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210093, China;
| | - Yi Shi
- Collaborative Innovation Center of Advanced Microstructures, School of Electronic Science and Engineering, Nanjing University, Nanjing 210093, China; (X.S.); (X.G.); (J.G.); (J.W.); (F.H.)
| | - Lijia Pan
- Collaborative Innovation Center of Advanced Microstructures, School of Electronic Science and Engineering, Nanjing University, Nanjing 210093, China; (X.S.); (X.G.); (J.G.); (J.W.); (F.H.)
| |
Collapse
|
47
|
Jin Z, Shi Y, Zhou L. Transparent sparse graph pathway network for analyzing the internal relationship of lung cancer. Front Genet 2024; 15:1437174. [PMID: 39411374 PMCID: PMC11473316 DOI: 10.3389/fgene.2024.1437174] [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: 05/27/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
While it is important to find the key biomarkers and improve the accuracy of disease models, it is equally important to understand their interaction relationships. In this study, a transparent sparse graph pathway network (TSGPN) is proposed based on the structure of graph neural networks. This network simulates the action of genes in vivo, adds to prior knowledge, and improves the model's accuracy. First, the graph connection was constructed according to protein-protein interaction networks and competing endogenous RNA (ceRNA) networks, from which some noise or unimportant connections were spontaneously removed based on the graph attention mechanism and hard concrete estimation. This realized the reconstruction of the ceRNA network representing the influence of other genes in the disease on mRNA. Next, the gene-based interpretation was transformed into a pathway-based interpretation based on the pathway database, and the hidden layer was added to realize the high-dimensional analysis of the pathway. Finally, the experimental results showed that the proposed TSGPN method is superior to other comparison methods in F1 score and AUC, and more importantly, it can effectively display the role of genes. Through data analysis applied to lung cancer prognosis, ten pathways related to LUSC prognosis were found, as well as the key biomarkers closely related to these pathways, such as HOXA10, hsa-mir-182, and LINC02544. The relationship between them was also reconstructed to better explain the internal mechanism of the disease.
Collapse
Affiliation(s)
- Zhibin Jin
- Information Engineering College, Shanghai Maritime University, pudong, China
| | - Yuhu Shi
- Information Engineering College, Shanghai Maritime University, pudong, China
| | - Lili Zhou
- Yangpu District Central Hospital, Shanghai, China
| |
Collapse
|
48
|
Zeng S, Adusumilli T, Awan SZ, Immadi MS, Xu D, Joshi T. G2PDeep-v2: a web-based deep-learning framework for phenotype prediction and biomarker discovery using multi-omics data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612292. [PMID: 39314346 PMCID: PMC11418982 DOI: 10.1101/2024.09.10.612292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The G2PDeep-v2 server is a web-based platform powered by deep learning, for phenotype prediction and markers discovery from multi-omics data in any organisms including humans, plants, animals, and viruses. The server provides multiple services for researchers to create deep-learning models through an interactive interface and train these models using an automated hyperparameter tuning algorithm on high-performance computing resources. Users can visualize the results of phenotype and markers predictions and perform Gene Set Enrichment Analysis for the significant markers to provide insights into the molecular mechanisms underlying complex diseases and other biological processes. The G2PDeep-v2 server is publicly available at https://g2pdeep.org/.
Collapse
Affiliation(s)
- Shuai Zeng
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA
| | - Trinath Adusumilli
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, 65211, USA
| | - Sania Zafar Awan
- MU Institute for Data Science and Informatics, University of Missouri-Columbia, Columbia, MO, 65211, USA
| | - Manish Sridhar Immadi
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, 65211, USA
| | - Dong Xu
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA
- MU Institute for Data Science and Informatics, University of Missouri-Columbia, Columbia, MO, 65211, USA
| | - Trupti Joshi
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA
- MU Institute for Data Science and Informatics, University of Missouri-Columbia, Columbia, MO, 65211, USA
- Department of Biomedical Informatics, Biostatistics and Medical Epidemiology, University of Missouri, Columbia, MO, 65211, USA
| |
Collapse
|
49
|
Yin Q, Hu Y, Dong Z, Lu J, Wang H. Cellular, Structural Basis, and Recent Progress for Targeting Murine Double Minute X (MDMX) in Tumors. J Med Chem 2024; 67:14723-14741. [PMID: 39185935 DOI: 10.1021/acs.jmedchem.4c00913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Murine double minute X (MDMX) is an oncoprotein that mainly has a negative regulatory effect on the tumor suppressor p53 to induce tumorigenesis. As MDMX is highly expressed in various types of tumor cells, targeting and inhibiting MDMX are becoming a promising strategy for treating cancers. However, the high degree of structural homology between MDMX and its homologous protein murine double minute 2 (MDM2) is a great challenge for the development of MDMX-targeted therapies. This review introduces the structure, distribution, and regulation of the MDMX, summarizes the structural features and structure-activity relationships (SARs) of MDMX ligands, and focuses on the differences between MDMX and MDM2 in these aspects. Our purpose of this work is to propose potential strategies to achieve the specific targeting of MDMX.
Collapse
Affiliation(s)
- Qikun Yin
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Yuemiao Hu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Zhiwen Dong
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Jing Lu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| |
Collapse
|
50
|
Qin T, Yang H, Wang L, Xue W, Yao N, Li Q, Chen X, Yang X, Yu X, Zhang Q, Li H. Molecule Design for Non-Aqueous Wide-Temperature Electrolytes via the Intelligentized Screening Method. Angew Chem Int Ed Engl 2024; 63:e202408902. [PMID: 38934230 DOI: 10.1002/anie.202408902] [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: 05/11/2024] [Revised: 06/06/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024]
Abstract
Operating a lithium-ion battery (LIB) in a wide temperature range is essential for ensuring a stable electricity supply amidst fluctuating temperatures caused by climate or terrain changes. Electrolyte plays a pivotal role in determining the temperature durability of batteries. However, specialized electrolytes designed for either low or high temperatures typically possess distinct features. Therefore, wide-temperature electrolytes (WTEs) are necessary as they encompass a combination of diverse properties, which complicates the clear instruction of WTE design. Here we represent an artificial intelligence (Al)-assisted workflow of WTE design through stepwise parameterizations and calculations. Linear mono-nitriles are identified as ideal wide-liquidus-range solvents that can "softly" solvate lithium ions by weak interactions. In addition, the explainable modules revealed the halogenoid similarity of cyanide as fluorine on the electrolyte properties (e.g. boiling point and dielectric constant). With the further introduction of an ether bond, 3-methoxypropionitrile (MPN) has been eventually determined as a main electrolyte solvent, enabling the battery operation from -60 to 120 °C. Particularly, a LiCoO2/Li cell using the proposed WTE can realize stable cycling with capacity retention reaching 72.3 % after 50 cycles under a high temperature of 100 °C.
Collapse
Affiliation(s)
- Tian Qin
- Beijing Frontier Research Center on Clean Energy, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haoyi Yang
- Beijing Frontier Research Center on Clean Energy, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Lei Wang
- Beijing Frontier Research Center on Clean Energy, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Weiran Xue
- Beijing Frontier Research Center on Clean Energy, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Nan Yao
- Beijing Key Laboratory of Green Chemical Reaction Engineering and Technology, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| | - Quan Li
- Beijing Frontier Research Center on Clean Energy, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xiang Chen
- Beijing Key Laboratory of Green Chemical Reaction Engineering and Technology, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| | - Xiukang Yang
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, National Base for International Science & Technology Cooperation, Hunan Province Key Laboratory for Electrochemical Energy Storage and Conversion, School of Chemistry, Xiangtan University, Hunan, Xiangtan, 411105, China
| | - Xiqian Yu
- Beijing Frontier Research Center on Clean Energy, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiang Zhang
- Beijing Key Laboratory of Green Chemical Reaction Engineering and Technology, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| | - Hong Li
- Beijing Frontier Research Center on Clean Energy, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|