1
|
Kumar R, Romano JD, Ritchie MD. Network-based analyses of multiomics data in biomedicine. BioData Min 2025; 18:37. [PMID: 40426270 PMCID: PMC12117783 DOI: 10.1186/s13040-025-00452-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
Network representations of data are designed to encode relationships between concepts as sets of edges between nodes. Human biology is inherently complex and is represented by data that often exists in a hierarchical nature. One canonical example is the relationship that exists within and between various -omics datasets, including genomics, transcriptomics, and proteomics, among others. Encoding such data in a network-based or graph-based representation allows the explicit incorporation of such relationships into various biomedical big data tasks, including (but not limited to) disease subtyping, interaction prediction, biomarker identification, and patient classification. This review will present various existing approaches in using network representations and analysis of data in multiomics in the framework of deep learning and machine learning approaches, subdivided into supervised and unsupervised approaches, to identify benefits and drawbacks of various approaches as well as the possible next steps for the field.
Collapse
Affiliation(s)
- Rachit Kumar
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Medical Scientist Training Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph D Romano
- Division of Informatics, Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marylyn D Ritchie
- Division of Informatics, Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Obeagu EI, Ezeanya CU, Ogenyi FC, Ifu DD. Big data analytics and machine learning in hematology: Transformative insights, applications and challenges. Medicine (Baltimore) 2025; 104:e41766. [PMID: 40068020 PMCID: PMC11902945 DOI: 10.1097/md.0000000000041766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/14/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
The integration of big data analytics and machine learning (ML) into hematology has ushered in a new era of precision medicine, offering transformative insights into disease management. By leveraging vast and diverse datasets, including genomic profiles, clinical laboratory results, and imaging data, these technologies enhance diagnostic accuracy, enable robust prognostic modeling, and support personalized therapeutic interventions. Advanced ML algorithms, such as neural networks and ensemble learning, facilitate the discovery of novel biomarkers and refine risk stratification for hematological disorders, including leukemias, lymphomas, and coagulopathies. Despite these advancements, significant challenges persist, particularly in the realms of data integration, algorithm validation, and ethical concerns. The heterogeneity of hematological datasets and the lack of standardized frameworks complicate their application, while the "black-box" nature of ML models raises issues of reliability and clinical trust. Moreover, safeguarding patient privacy in an era of data-driven medicine remains paramount, necessitating the development of secure and ethical analytical practices. Addressing these challenges is critical to ensuring equitable and effective implementation of these technologies. Collaborative efforts between hematologists, data scientists, and bioinformaticians are pivotal in translating these innovations into real-world clinical practice. Emphasis on developing explainable artificial intelligence models, integrating real-time analytics, and adopting federated learning approaches will further enhance the utility and adoption of these technologies. As big data analytics and ML continue to evolve, their potential to revolutionize hematology and improve patient outcomes remains immense.
Collapse
Affiliation(s)
| | | | - Fabian Chukwudi Ogenyi
- Department of Electrical, Telecommunication and Computer Engineering, Kampala International University, Kampala, Uganda
| | - Deborah Domini Ifu
- Department of Biomedical and Laboratory Science, Africa University, Mutare, Zimbabwe
| |
Collapse
|
3
|
Kan Y, Qi Y, Zhang Z, Liang X, Wang W, Jin S. Integration of unpaired single cell omics data by deep transfer graph convolutional network. PLoS Comput Biol 2025; 21:e1012625. [PMID: 39821189 PMCID: PMC11778791 DOI: 10.1371/journal.pcbi.1012625] [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: 02/02/2024] [Revised: 01/29/2025] [Accepted: 11/09/2024] [Indexed: 01/19/2025] Open
Abstract
The rapid advance of large-scale atlas-level single cell RNA sequences and single-cell chromatin accessibility data provide extraordinary avenues to broad and deep insight into complex biological mechanism. Leveraging the datasets and transfering labels from scRNA-seq to scATAC-seq will empower the exploration of single-cell omics data. However, the current label transfer methods have limited performance, largely due to the lower capable of preserving fine-grained cell populations and intrinsic or extrinsic heterogeneity between datasets. Here, we present a robust deep transfer model based graph convolutional network, scTGCN, which achieves versatile performance in preserving biological variation, while achieving integration hundreds of thousands cells in minutes with low memory consumption. We show that scTGCN is powerful to the integration of mouse atlas data and multimodal data generated from APSA-seq and CITE-seq. Thus, scTGCN shows high label transfer accuracy and effectively knowledge transfer across different modalities.
Collapse
Affiliation(s)
- Yulong Kan
- School of Mathematics/Harbin Institute of Technology, Harbin, China
| | - Yunjing Qi
- School of Mathematics/Harbin Institute of Technology, Harbin, China
| | - Zhongxiao Zhang
- School of Mathematics/Harbin Institute of Technology, Harbin, China
| | - Xikeng Liang
- School of Mathematics/Harbin Institute of Technology, Harbin, China
| | - Weihao Wang
- School of Mathematics/Harbin Institute of Technology, Harbin, China
| | - Shuilin Jin
- School of Mathematics/Harbin Institute of Technology, Harbin, China
| |
Collapse
|
4
|
Lee MA, Vyas P, D'Agostino F, Wieben A, Coviak C, Mullen-Fortino M, Park S, Sileo M, Nogueira de Souza E, Brown S, Role J, Reger A, Pruinelli L. Empowering Nurses Through Data Literacy and Data Science Literacy: Insights From a State-of-the-Art Literature Review. ANS Adv Nurs Sci 2024:00012272-990000000-00100. [PMID: 39356110 DOI: 10.1097/ans.0000000000000546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Affiliation(s)
- Mikyoung Angela Lee
- Author Affiliations: Texas Woman's University, Dallas, Texas (Dr Lee); University of Arizona, Tucson, Arizona (Mr Vyas); Saint Camillus International University of Health Sciences, Rome, Italy (Dr D'Agostino); University of Wisconsin-Madison, Madison, Wisconsin (Dr Wieben); Grand Valley State University, Allendale, Michigan (Dr Coviak); Penn Presbyterian Medical Center, Philadelphia, Pennsylvania (Dr Mullen-Fortino); University of Minnesota, Minneapolis, Minnesota (Ms Park); Independent contributor, Boston, Massachusetts (Ms Sileo); Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil (Dr Nogueira de Souza); Memorial Sloan Kettering Cancer Center, New York, New York (Dr Brown); Loma Linda University Health, Loma Linda, California (Dr Role); Independent Contributor, St Louis, Missouri (Dr Reger); and University of Florida, Gainesville, Florida (Dr Pruinelli)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Ruprecht NA, Kennedy JD, Bansal B, Singhal S, Sens D, Maggio A, Doe V, Hawkins D, Campbel R, O’Connell K, Gill JS, Schaefer K, Singhal SK. Transcriptomics and epigenetic data integration learning module on Google Cloud. Brief Bioinform 2024; 25:bbae352. [PMID: 39101486 PMCID: PMC11299028 DOI: 10.1093/bib/bbae352] [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: 11/30/2023] [Revised: 05/12/2024] [Accepted: 07/06/2024] [Indexed: 08/06/2024] Open
Abstract
Multi-omics (genomics, transcriptomics, epigenomics, proteomics, metabolomics, etc.) research approaches are vital for understanding the hierarchical complexity of human biology and have proven to be extremely valuable in cancer research and precision medicine. Emerging scientific advances in recent years have made high-throughput genome-wide sequencing a central focus in molecular research by allowing for the collective analysis of various kinds of molecular biological data from different types of specimens in a single tissue or even at the level of a single cell. Additionally, with the help of improved computational resources and data mining, researchers are able to integrate data from different multi-omics regimes to identify new prognostic, diagnostic, or predictive biomarkers, uncover novel therapeutic targets, and develop more personalized treatment protocols for patients. For the research community to parse the scientifically and clinically meaningful information out of all the biological data being generated each day more efficiently with less wasted resources, being familiar with and comfortable using advanced analytical tools, such as Google Cloud Platform becomes imperative. This project is an interdisciplinary, cross-organizational effort to provide a guided learning module for integrating transcriptomics and epigenetics data analysis protocols into a comprehensive analysis pipeline for users to implement in their own work, utilizing the cloud computing infrastructure on Google Cloud. The learning module consists of three submodules that guide the user through tutorial examples that illustrate the analysis of RNA-sequence and Reduced-Representation Bisulfite Sequencing data. The examples are in the form of breast cancer case studies, and the data sets were procured from the public repository Gene Expression Omnibus. The first submodule is devoted to transcriptomics analysis with the RNA sequencing data, the second submodule focuses on epigenetics analysis using the DNA methylation data, and the third submodule integrates the two methods for a deeper biological understanding. The modules begin with data collection and preprocessing, with further downstream analysis performed in a Vertex AI Jupyter notebook instance with an R kernel. Analysis results are returned to Google Cloud buckets for storage and visualization, removing the computational strain from local resources. The final product is a start-to-finish tutorial for the researchers with limited experience in multi-omics to integrate transcriptomics and epigenetics data analysis into a comprehensive pipeline to perform their own biological research.This manuscript describes the development of a resource module that is part of a learning platform named ``NIGMS Sandbox for Cloud-based Learning'' https://github.com/NIGMS/NIGMS-Sandbox. The overall genesis of the Sandbox is described in the editorial NIGMS Sandbox [16] at the beginning of this Supplement. This module delivers learning materials on the analysis of bulk and single-cell ATAC-seq data in an interactive format that uses appropriate cloud resources for data access and analyses. HIGHLIGHTS
Collapse
Affiliation(s)
- Nathan A Ruprecht
- Department of Biomedical Engineering, University of North Dakota, 501 N. Columbia Road Stop 8380, Grand Forks, ND 58202, United States
| | - Joshua D Kennedy
- Department of Biomedical Engineering, University of North Dakota, 501 N. Columbia Road Stop 8380, Grand Forks, ND 58202, United States
- Department of Chemistry and Physics, Drury University, 900 N. Benton Avenue, Springfield, MO 65802, United States
| | - Benu Bansal
- Department of Biomedical Engineering, University of North Dakota, 501 N. Columbia Road Stop 8380, Grand Forks, ND 58202, United States
| | - Sonalika Singhal
- Department of Pathology, University of North Dakota, 1301 N. Columbia Road Stop 9037, Grand Forks, ND 58202, United States
| | - Donald Sens
- Department of Pathology, University of North Dakota, 1301 N. Columbia Road Stop 9037, Grand Forks, ND 58202, United States
| | - Angela Maggio
- Deloitte, Health Data and AI, Deloitte Consulting LLP, 1919 N. Lynn Street, Suite 1500, Arlington, VA 22209, United States
| | - Valena Doe
- Google, Google Cloud, 1900 Reston Metro Plaza, Reston, VA 20190, United States
| | - Dale Hawkins
- Google, Google Cloud, 1900 Reston Metro Plaza, Reston, VA 20190, United States
| | - Ross Campbel
- NIH Center for Information Technology (CIT), 6555 Rock Spring Drive, Bethesda, MD 20892, United States
| | - Kyle O’Connell
- NIH Center for Information Technology (CIT), 6555 Rock Spring Drive, Bethesda, MD 20892, United States
| | - Jappreet Singh Gill
- Department of Biomedical Engineering, University of North Dakota, 501 N. Columbia Road Stop 8380, Grand Forks, ND 58202, United States
| | - Kalli Schaefer
- Department of Biomedical Engineering, University of North Dakota, 501 N. Columbia Road Stop 8380, Grand Forks, ND 58202, United States
| | - Sandeep K Singhal
- Department of Biomedical Engineering, University of North Dakota, 501 N. Columbia Road Stop 8380, Grand Forks, ND 58202, United States
- Department of Pathology, University of North Dakota, 1301 N. Columbia Road Stop 9037, Grand Forks, ND 58202, United States
| |
Collapse
|
6
|
Yuan J, Mi L, Wang S, Cheng Y, Hou X. Comparing the influence of big data resources on medical knowledge recall for staff with and without medical collaboration platform. BMC MEDICAL EDUCATION 2023; 23:956. [PMID: 38093304 PMCID: PMC10720120 DOI: 10.1186/s12909-023-04926-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND This study aims to examine how big data resources affect the recall of prior medical knowledge by healthcare professionals, and how this differs in environments with and without remote consultation platforms. METHOD This study investigated two distinct categories of medical institutions, namely 132 medical institutions with platforms, and 176 medical institutions without the platforms. Big data resources are categorized into two levels-medical institutional level and public level-and three types, namely data, technology, and services. The data are analyzed using SmartPLS2. RESULTS (1) In both scenarios, shared big data resources at the public level have a significant direct impact on the recall of prior medical knowledge. However, there is a significant difference in the direct impact of big data resources at the institutional level in both scenarios. (2) In institutions with platforms, for the three big data resources (the medical big data assets and big data deployment technical capacity at the medical institutional level, and policies of medical big data at the public level) without direct impacts, there exist three indirect pathways. (3) In institutions without platforms, for the two big data resources (the service capability and big data technical capacity at the medical institutional level) without direct impacts, there exist three indirect pathways. CONCLUSIONS The different interactions between big data, technology, and services, as well as between different levels of big data resources, affect the way clinical doctors recall relevant medical knowledge. These interaction patterns vary between institutions with and without platforms. This study provides a reference for governments and institutions to design big data environments for improving clinical capabilities.
Collapse
Affiliation(s)
- JunYi Yuan
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, China
| | - Linhui Mi
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, China
| | - SuFen Wang
- Glorious Sun School of Business and Management, Donghua University, 1882 West Yanan Road, Shanghai, China
| | - Yuejia Cheng
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, China
| | - Xumin Hou
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, China.
| |
Collapse
|
7
|
Feuerecker B, Heimer MM, Geyer T, Fabritius MP, Gu S, Schachtner B, Beyer L, Ricke J, Gatidis S, Ingrisch M, Cyran CC. Artificial Intelligence in Oncological Hybrid Imaging. Nuklearmedizin 2023; 62:296-305. [PMID: 37802057 DOI: 10.1055/a-2157-6810] [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: 10/08/2023]
Abstract
BACKGROUND Artificial intelligence (AI) applications have become increasingly relevant across a broad spectrum of settings in medical imaging. Due to the large amount of imaging data that is generated in oncological hybrid imaging, AI applications are desirable for lesion detection and characterization in primary staging, therapy monitoring, and recurrence detection. Given the rapid developments in machine learning (ML) and deep learning (DL) methods, the role of AI will have significant impact on the imaging workflow and will eventually improve clinical decision making and outcomes. METHODS AND RESULTS The first part of this narrative review discusses current research with an introduction to artificial intelligence in oncological hybrid imaging and key concepts in data science. The second part reviews relevant examples with a focus on applications in oncology as well as discussion of challenges and current limitations. CONCLUSION AI applications have the potential to leverage the diagnostic data stream with high efficiency and depth to facilitate automated lesion detection, characterization, and therapy monitoring to ultimately improve quality and efficiency throughout the medical imaging workflow. The goal is to generate reproducible, structured, quantitative diagnostic data for evidence-based therapy guidance in oncology. However, significant challenges remain regarding application development, benchmarking, and clinical implementation. KEY POINTS · Hybrid imaging generates a large amount of multimodality medical imaging data with high complexity and depth.. · Advanced tools are required to enable fast and cost-efficient processing along the whole radiology value chain.. · AI applications promise to facilitate the assessment of oncological disease in hybrid imaging with high quality and efficiency for lesion detection, characterization, and response assessment. The goal is to generate reproducible, structured, quantitative diagnostic data for evidence-based oncological therapy guidance.. · Selected applications in three oncological entities (lung, prostate, and neuroendocrine tumors) demonstrate how AI algorithms may impact imaging-based tasks in hybrid imaging and potentially guide clinical decision making..
Collapse
Affiliation(s)
- Benedikt Feuerecker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Partner site Munich, DKTK German Cancer Consortium, Munich, Germany
| | - Maurice M Heimer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Thomas Geyer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Sijing Gu
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Sergios Gatidis
- Department of Radiology, University Hospital Tübingen, Tübingen, Germany
- MPI, Max Planck Institute for Intelligent Systems, Tübingen, Germany
| | - Michael Ingrisch
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Clemens C Cyran
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
8
|
Tang Z, Li Z, Hou T, Zhang T, Yang B, Su J, Song Q. SiGra: single-cell spatial elucidation through an image-augmented graph transformer. Nat Commun 2023; 14:5618. [PMID: 37699885 PMCID: PMC10497630 DOI: 10.1038/s41467-023-41437-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 09/01/2023] [Indexed: 09/14/2023] Open
Abstract
Recent advances in high-throughput molecular imaging have pushed spatial transcriptomics technologies to subcellular resolution, which surpasses the limitations of both single-cell RNA-seq and array-based spatial profiling. The multichannel immunohistochemistry images in such data provide rich information on the cell types, functions, and morphologies of cellular compartments. In this work, we developed a method, single-cell spatial elucidation through image-augmented Graph transformer (SiGra), to leverage such imaging information for revealing spatial domains and enhancing substantially sparse and noisy transcriptomics data. SiGra applies hybrid graph transformers over a single-cell spatial graph. SiGra outperforms state-of-the-art methods on both single-cell and spot-level spatial transcriptomics data from complex tissues. The inclusion of immunohistochemistry images improves the model performance by 37% (95% CI: 27-50%). SiGra improves the characterization of intratumor heterogeneity and intercellular communication and recovers the known microscopic anatomy. Overall, SiGra effectively integrates different spatial modality data to gain deep insights into spatial cellular ecosystems.
Collapse
Affiliation(s)
- Ziyang Tang
- Department of Computer and Information Technology, Purdue University, Indiana, USA
| | - Zuotian Li
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana, USA
- Department of Computer Graphics Technology, Purdue University, Indiana, USA
| | - Tieying Hou
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indiana, USA
| | - Tonglin Zhang
- Department of Statistics, Purdue University, Indiana, USA
| | - Baijian Yang
- Department of Computer and Information Technology, Purdue University, Indiana, USA.
| | - Jing Su
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana, USA.
| | - Qianqian Song
- Department of Cancer Biology, Wake Forest University School of Medicine, North Carolina, USA.
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Florida, USA.
| |
Collapse
|
9
|
Feuerecker B, Heimer MM, Geyer T, Fabritius MP, Gu S, Schachtner B, Beyer L, Ricke J, Gatidis S, Ingrisch M, Cyran CC. Artificial Intelligence in Oncological Hybrid Imaging. ROFO-FORTSCHR RONTG 2023; 195:105-114. [PMID: 36170852 DOI: 10.1055/a-1909-7013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Artificial intelligence (AI) applications have become increasingly relevant across a broad spectrum of settings in medical imaging. Due to the large amount of imaging data that is generated in oncological hybrid imaging, AI applications are desirable for lesion detection and characterization in primary staging, therapy monitoring, and recurrence detection. Given the rapid developments in machine learning (ML) and deep learning (DL) methods, the role of AI will have significant impact on the imaging workflow and will eventually improve clinical decision making and outcomes. METHODS AND RESULTS The first part of this narrative review discusses current research with an introduction to artificial intelligence in oncological hybrid imaging and key concepts in data science. The second part reviews relevant examples with a focus on applications in oncology as well as discussion of challenges and current limitations. CONCLUSION AI applications have the potential to leverage the diagnostic data stream with high efficiency and depth to facilitate automated lesion detection, characterization, and therapy monitoring to ultimately improve quality and efficiency throughout the medical imaging workflow. The goal is to generate reproducible, structured, quantitative diagnostic data for evidence-based therapy guidance in oncology. However, significant challenges remain regarding application development, benchmarking, and clinical implementation. KEY POINTS · Hybrid imaging generates a large amount of multimodality medical imaging data with high complexity and depth.. · Advanced tools are required to enable fast and cost-efficient processing along the whole radiology value chain.. · AI applications promise to facilitate the assessment of oncological disease in hybrid imaging with high quality and efficiency for lesion detection, characterization, and response assessment. The goal is to generate reproducible, structured, quantitative diagnostic data for evidence-based oncological therapy guidance.. · Selected applications in three oncological entities (lung, prostate, and neuroendocrine tumors) demonstrate how AI algorithms may impact imaging-based tasks in hybrid imaging and potentially guide clinical decision making.. CITATION FORMAT · Feuerecker B, Heimer M, Geyer T et al. Artificial Intelligence in Oncological Hybrid Imaging. Fortschr Röntgenstr 2023; 195: 105 - 114.
Collapse
Affiliation(s)
- Benedikt Feuerecker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany.,German Cancer Research Center (DKFZ), Partner site Munich, DKTK German Cancer Consortium, Munich, Germany
| | - Maurice M Heimer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Thomas Geyer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Sijing Gu
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Sergios Gatidis
- Department of Radiology, University Hospital Tübingen, Tübingen, Germany.,MPI, Max Planck Institute for Intelligent Systems, Tübingen, Germany
| | - Michael Ingrisch
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Clemens C Cyran
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
10
|
Goodwin AJ, Eytan D, Dixon W, Goodfellow SD, Doherty Z, Greer RW, McEwan A, Tracy M, Laussen PC, Assadi A, Mazwi M. Timing errors and temporal uncertainty in clinical databases-A narrative review. Front Digit Health 2022; 4:932599. [PMID: 36060541 PMCID: PMC9433547 DOI: 10.3389/fdgth.2022.932599] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/11/2022] [Indexed: 11/28/2022] Open
Abstract
A firm concept of time is essential for establishing causality in a clinical setting. Review of critical incidents and generation of study hypotheses require a robust understanding of the sequence of events but conducting such work can be problematic when timestamps are recorded by independent and unsynchronized clocks. Most clinical models implicitly assume that timestamps have been measured accurately and precisely, but this custom will need to be re-evaluated if our algorithms and models are to make meaningful use of higher frequency physiological data sources. In this narrative review we explore factors that can result in timestamps being erroneously recorded in a clinical setting, with particular focus on systems that may be present in a critical care unit. We discuss how clocks, medical devices, data storage systems, algorithmic effects, human factors, and other external systems may affect the accuracy and precision of recorded timestamps. The concept of temporal uncertainty is introduced, and a holistic approach to timing accuracy, precision, and uncertainty is proposed. This quantitative approach to modeling temporal uncertainty provides a basis to achieve enhanced model generalizability and improved analytical outcomes.
Collapse
Affiliation(s)
- Andrew J. Goodwin
- Department of Critical Care Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- School of Biomedical Engineering, University of Sydney, Sydney, NSW, Australia
| | - Danny Eytan
- Department of Critical Care Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - William Dixon
- Department of Critical Care Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sebastian D. Goodfellow
- Department of Critical Care Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Civil and Mineral Engineering, University of Toronto, Toronto, ON, Canada
| | - Zakary Doherty
- Research Fellow, School of Rural Health, Monash University, Melbourne, VIC, Australia
| | - Robert W. Greer
- Department of Critical Care Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Alistair McEwan
- School of Biomedical Engineering, University of Sydney, Sydney, NSW, Australia
| | - Mark Tracy
- Neonatal Intensive Care Unit, Westmead Hospital, Sydney, NSW, Australia
- Department of Paediatrics and Child Health, The University of Sydney, Sydney, NSW, Australia
| | - Peter C. Laussen
- Department of Anesthesia, Boston Children's Hospital, Boston, MA, United States
| | - Azadeh Assadi
- Department of Critical Care Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Engineering and Applied Sciences, Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Mjaye Mazwi
- Department of Critical Care Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Shen X, Shen H, Wu D, Feng M, Hu J, Liu J, Yang Y, Yang M, Li Y, Shi L, Chen K, Li X. Scalable batch-correction approach for integrating large-scale single-cell transcriptomes. Brief Bioinform 2022; 23:6659742. [PMID: 35947966 DOI: 10.1093/bib/bbac327] [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: 04/30/2022] [Revised: 06/26/2022] [Accepted: 07/19/2022] [Indexed: 11/14/2022] Open
Abstract
Integration of accumulative large-scale single-cell transcriptomes requires scalable batch-correction approaches. Here we propose Fugue, a simple and efficient batch-correction method that is scalable for integrating super large-scale single-cell transcriptomes from diverse sources. The core idea of the method is to encode batch information as trainable parameters and add it to single-cell expression profile; subsequently, a contrastive learning approach is used to learn feature representation of the additive expression profile. We demonstrate the scalability of Fugue by integrating all single cells obtained from the Human Cell Atlas. We benchmark Fugue against current state-of-the-art methods and show that Fugue consistently achieves improved performance in terms of data alignment and clustering preservation. Our study will facilitate the integration of single-cell transcriptomes at increasingly large scale.
Collapse
Affiliation(s)
- Xilin Shen
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Hongru Shen
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Dan Wu
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Mengyao Feng
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Jiani Hu
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Jilei Liu
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Yichen Yang
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Meng Yang
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Yang Li
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Lei Shi
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Xiangchun Li
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
12
|
Wang S, Yuan J, Pan C. Impact of big data resources on clinicians’ activation of prior medical knowledge. Heliyon 2022; 8:e10312. [PMID: 36105474 PMCID: PMC9465108 DOI: 10.1016/j.heliyon.2022.e10312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 07/10/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022] Open
Abstract
Background Activating prior medical knowledge in diagnosis and treatment is an important basis for clinicians to improve their care ability. However, it has not been systematically explained whether and how various big data resources affect the activation of prior knowledge in the big data environment faced by clinicians. Objective The aim of this study is to contribute to a better understanding on how the activation of prior knowledge of clinicians is affected by a wide range of shared and private big data resources, to reveal the impact of big data resources on clinical competence and professional development of clinicians. Method Through the comprehensive analysis of extant research results, big data resources are classified as big data itself, big data technology and big data services at the public and institutional levels. A survey was conducted on clinicians and IT personnel in Chinese hospitals. A total of 616 surveys are completed, involving 308 medical institutions. Each medical institution includes a clinician and an IT personnel. SmartPLS version 2.0 software package was used to test the direct impact of big data resources on the activation of prior knowledge. We further analyze their indirect impact of those big data resources without direct impact. Results (1) Big data quality environment at the institutional level and the big data sharing environment at the public level directly affect activation of prior medical knowledge; (2) Big data service environment at the institutional level directly affects activation of prior medical knowledge; (3) Big data deployment environment at the institutional level and big data service environment at the public level have no direct impact on activation of prior knowledge of clinicians, but they have an indirect impact through big data quality environment and service environment at the institutional level and the big data sharing environment at the public level. Conclusions Big data technology, big data itself and big data service at the public level and institutional level interact and influence each other to activate prior medical knowledge. This study highlights the implications of big data resources on improvement of clinicians’ diagnosis and treatment ability.
Collapse
Affiliation(s)
- Sufen Wang
- Glorious Sun School of Business and Management, DongHua University, Shanghai, China
| | - Junyi Yuan
- Information Center, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
- Corresponding author.
| | - Changqing Pan
- Hospital's Office, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
13
|
Eva G, Liese G, Stephanie B, Petr H, Leslie M, Roel V, Martine V, Sergi B, Mette H, Sarah J, Laura RM, Arnout S, Morris A S, Jan T, Xenia T, Nina V, Koert VE, Sylvie R, Greet S. Position paper on management of personal data in environment and health research in Europe. ENVIRONMENT INTERNATIONAL 2022; 165:107334. [PMID: 35696847 DOI: 10.1016/j.envint.2022.107334] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
Management of datasets that include health information and other sensitive personal information of European study participants has to be compliant with the General Data Protection Regulation (GDPR, Regulation (EU) 2016/679). Within scientific research, the widely subscribed'FAIR' data principles should apply, meaning that research data should be findable, accessible, interoperable and re-usable. Balancing the aim of open science driven FAIR data management with GDPR compliant personal data protection safeguards is now a common challenge for many research projects dealing with (sensitive) personal data. In December 2020 a workshop was held with representatives of several large EU research consortia and of the European Commission to reflect on how to apply the FAIR data principles for environment and health research (E&H). Several recent data intensive EU funded E&H research projects face this challenge and work intensively towards developing solutions to access, exchange, store, handle, share, process and use such sensitive personal data, with the aim to support European and transnational collaborations. As a result, several recommendations, opportunities and current limitations were formulated. New technical developments such as federated data management and analysis systems, machine learning together with advanced search software, harmonized ontologies and data quality standards should in principle facilitate the FAIRification of data. To address ethical, legal, political and financial obstacles to the wider re-use of data for research purposes, both specific expertise and underpinning infrastructure are needed. There is a need for the E&H research data to find their place in the European Open Science Cloud. Communities using health and population data, environmental data and other publicly available data have to interconnect and synergize. To maximize the use and re-use of environment and health data, a dedicated supporting European infrastructure effort, such as the EIRENE research infrastructure within the ESFRI roadmap 2021, is needed that would interact with existing infrastructures.
Collapse
Affiliation(s)
- Govarts Eva
- VITO Health, Flemish Institute for Technological Research (VITO), Mol, Belgium.
| | - Gilles Liese
- VITO Health, Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Bopp Stephanie
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | - Matalonga Leslie
- CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Vermeulen Roel
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
| | - Vrijheid Martine
- ISGlobal, Barcelona, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Beltran Sergi
- CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, Spain
| | - Hartlev Mette
- Faculty of Law, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Standaert Arnout
- VITO Health, Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Swertz Morris A
- Department of Genetics & Genomics Coordination Center, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Theunis Jan
- VITO Health, Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Trier Xenia
- European Environment Agency (EEA), Copenhagen, Denmark
| | - Vogel Nina
- German Environment Agency (UBA), Berlin, Germany
| | | | - Remy Sylvie
- VITO Health, Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Schoeters Greet
- VITO Health, Flemish Institute for Technological Research (VITO), Mol, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
14
|
Zhao J, Li Q. Big Data-Artificial Intelligence Fusion Technology in Education in the Context of the New Crown Epidemic. BIG DATA 2022; 10:262-276. [PMID: 35605025 DOI: 10.1089/big.2021.0245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
This article presents an in-depth study and analysis of the application of big data-artificial intelligence fusion technology to the field of education in the context of the New Crown epidemic. Since the outbreak of the New Crown epidemic, there is a need to understand the channels for solving the demands of productive life during the epidemic, and the main way to solve these problems is to apply the Internet, big data, and artificial intelligence. Therefore, exploring the application of big data-artificial intelligence fusion technology in education in the context of the New Crest pneumonia epidemic is a top priority for reform and development nowadays. This study uses the paradigm narrow shift analysis framework to verify whether Computer Aided Instructional design, multimedia instructional design, and informational instructional design produce migration. For the intelligent stage of instructional design, the inevitability of the change in basic assumptions of instructional design in the context of artificial intelligence (AI) is first explained in terms of the opportunities brought by AI to education and teaching, the problems of the original information-based instructional design itself, and the many challenges it faces. On this basis, we also use the change in basic assumptions analysis framework to explain the content of intelligent instructional design by using the four elements of beliefs, values, symbols, and paradigms promised by the members of the community, verify that it has shifted, and build a change in the basic assumptions model from multimedia instructional design to information-based instructional design to intelligent instructional design. The article gives three countermeasures to solve the problem, that is, raising awareness, improving the plan, and strengthening the drill. To ensure the smooth implementation of the emergency management of national covid control program (NCCP), higher education institutions should further strengthen the construction efforts of specialized psychological counseling teams, build an early warning mechanism for psychological problems of the NCCP epidemic in higher education institutions, and a multilevel supervision system.
Collapse
Affiliation(s)
- JunJing Zhao
- School of Economics and Finance, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qi Li
- School of Economics and Finance, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
15
|
Charow R, Jeyakumar T, Younus S, Dolatabadi E, Salhia M, Al-Mouaswas D, Anderson M, Balakumar S, Clare M, Dhalla A, Gillan C, Haghzare S, Jackson E, Lalani N, Mattson J, Peteanu W, Tripp T, Waldorf J, Williams S, Tavares W, Wiljer D. Artificial Intelligence Education Programs for Health Care Professionals: Scoping Review. JMIR MEDICAL EDUCATION 2021; 7:e31043. [PMID: 34898458 PMCID: PMC8713099 DOI: 10.2196/31043] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 05/12/2023]
Abstract
BACKGROUND As the adoption of artificial intelligence (AI) in health care increases, it will become increasingly crucial to involve health care professionals (HCPs) in developing, validating, and implementing AI-enabled technologies. However, because of a lack of AI literacy, most HCPs are not adequately prepared for this revolution. This is a significant barrier to adopting and implementing AI that will affect patients. In addition, the limited existing AI education programs face barriers to development and implementation at various levels of medical education. OBJECTIVE With a view to informing future AI education programs for HCPs, this scoping review aims to provide an overview of the types of current or past AI education programs that pertains to the programs' curricular content, modes of delivery, critical implementation factors for education delivery, and outcomes used to assess the programs' effectiveness. METHODS After the creation of a search strategy and keyword searches, a 2-stage screening process was conducted by 2 independent reviewers to determine study eligibility. When consensus was not reached, the conflict was resolved by consulting a third reviewer. This process consisted of a title and abstract scan and a full-text review. The articles were included if they discussed an actual training program or educational intervention, or a potential training program or educational intervention and the desired content to be covered, focused on AI, and were designed or intended for HCPs (at any stage of their career). RESULTS Of the 10,094 unique citations scanned, 41 (0.41%) studies relevant to our eligibility criteria were identified. Among the 41 included studies, 10 (24%) described 13 unique programs and 31 (76%) discussed recommended curricular content. The curricular content of the unique programs ranged from AI use, AI interpretation, and cultivating skills to explain results derived from AI algorithms. The curricular topics were categorized into three main domains: cognitive, psychomotor, and affective. CONCLUSIONS This review provides an overview of the current landscape of AI in medical education and highlights the skills and competencies required by HCPs to effectively use AI in enhancing the quality of care and optimizing patient outcomes. Future education efforts should focus on the development of regulatory strategies, a multidisciplinary approach to curriculum redesign, a competency-based curriculum, and patient-clinician interaction.
Collapse
Affiliation(s)
- Rebecca Charow
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- University Health Network, Toronto, ON, Canada
| | | | | | - Elham Dolatabadi
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Vector Institute, Toronto, ON, Canada
| | - Mohammad Salhia
- Michener Institute of Education, University Health Network, Toronto, ON, Canada
| | - Dalia Al-Mouaswas
- Michener Institute of Education, University Health Network, Toronto, ON, Canada
| | | | - Sarmini Balakumar
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Michener Institute of Education, University Health Network, Toronto, ON, Canada
| | - Megan Clare
- Michener Institute of Education, University Health Network, Toronto, ON, Canada
| | | | - Caitlin Gillan
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- University Health Network, Toronto, ON, Canada
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Shabnam Haghzare
- University Health Network, Toronto, ON, Canada
- Vector Institute, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | | | | | - Jane Mattson
- Michener Institute of Education, University Health Network, Toronto, ON, Canada
| | - Wanda Peteanu
- Michener Institute of Education, University Health Network, Toronto, ON, Canada
| | - Tim Tripp
- University Health Network, Toronto, ON, Canada
| | - Jacqueline Waldorf
- Michener Institute of Education, University Health Network, Toronto, ON, Canada
| | | | - Walter Tavares
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- University Health Network, Toronto, ON, Canada
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Wilson Centre, Toronto, ON, Canada
| | - David Wiljer
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- University Health Network, Toronto, ON, Canada
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- CAMH Education, Centre for Addictions and Mental Health (CAMH), Toronto, ON, Canada
| |
Collapse
|
16
|
Song Q, Su J. DSTG: deconvoluting spatial transcriptomics data through graph-based artificial intelligence. Brief Bioinform 2021; 22:bbaa414. [PMID: 33480403 PMCID: PMC8425268 DOI: 10.1093/bib/bbaa414] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/08/2020] [Accepted: 12/15/2020] [Indexed: 12/30/2022] Open
Abstract
Recent development of spatial transcriptomics (ST) is capable of associating spatial information at different spots in the tissue section with RNA abundance of cells within each spot, which is particularly important to understand tissue cytoarchitectures and functions. However, for such ST data, since a spot is usually larger than an individual cell, gene expressions measured at each spot are from a mixture of cells with heterogenous cell types. Therefore, ST data at each spot needs to be disentangled so as to reveal the cell compositions at that spatial spot. In this study, we propose a novel method, named deconvoluting spatial transcriptomics data through graph-based convolutional networks (DSTG), to accurately deconvolute the observed gene expressions at each spot and recover its cell constitutions, thus achieving high-level segmentation and revealing spatial architecture of cellular heterogeneity within tissues. DSTG not only demonstrates superior performance on synthetic spatial data generated from different protocols, but also effectively identifies spatial compositions of cells in mouse cortex layer, hippocampus slice and pancreatic tumor tissues. In conclusion, DSTG accurately uncovers the cell states and subpopulations based on spatial localization. DSTG is available as a ready-to-use open source software (https://github.com/Su-informatics-lab/DSTG) for precise interrogation of spatial organizations and functions in tissues.
Collapse
Affiliation(s)
- Qianqian Song
- Center for Cancer Genomics and Precision Oncology, Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston Salem, NC 27157, USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Jing Su
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
17
|
Song Q, Su J, Zhang W. scGCN is a graph convolutional networks algorithm for knowledge transfer in single cell omics. Nat Commun 2021; 12:3826. [PMID: 34158507 PMCID: PMC8219725 DOI: 10.1038/s41467-021-24172-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 06/07/2021] [Indexed: 12/20/2022] Open
Abstract
Single-cell omics is the fastest-growing type of genomics data in the literature and public genomics repositories. Leveraging the growing repository of labeled datasets and transferring labels from existing datasets to newly generated datasets will empower the exploration of single-cell omics data. However, the current label transfer methods have limited performance, largely due to the intrinsic heterogeneity among cell populations and extrinsic differences between datasets. Here, we present a robust graph artificial intelligence model, single-cell Graph Convolutional Network (scGCN), to achieve effective knowledge transfer across disparate datasets. Through benchmarking with other label transfer methods on a total of 30 single cell omics datasets, scGCN consistently demonstrates superior accuracy on leveraging cells from different tissues, platforms, and species, as well as cells profiled at different molecular layers. scGCN is implemented as an integrated workflow as a python software, which is available at https://github.com/QSong-github/scGCN .
Collapse
Affiliation(s)
- Qianqian Song
- Center for Cancer Genomics and Precision Oncology, Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston Salem, NC, USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Jing Su
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA.
- Section on Gerontology and Geriatric Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | - Wei Zhang
- Center for Cancer Genomics and Precision Oncology, Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston Salem, NC, USA.
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, USA.
| |
Collapse
|
18
|
Diwadkar AR, Yoon S, Shim J, Gonzalez M, Urbanowicz R, Himes BE. Integrating Biomedical Informatics Training into Existing High School Curricula. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2021; 2021:190-199. [PMID: 34457133 PMCID: PMC8378629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Growing demand for biomedical informaticists and expertise in areas related to this discipline has accentuated the need to integrate biomedical informatics training into high school curricula. The K-12 Bioinformatics professional development project educates high school teachers about data analysis, biomedical informatics and mobile learning, and partners with them to expose high school students to health and environment-related issues using biomedical informatics knowledge and current technologies. We designed low-cost pollution sensors and created interactive web applications that teachers from six Philadelphia public high schools used during the 2019-2020 school year to successfully implement a problem-based mobile learning unit that included collecting and interpreting air pollution data, as well as relating this data to asthma. Through this project, we sought to improve data and health literacy among the students and teachers, while inspiring student engagement by demonstrating how biomedical informatics can help address problems relevant to communities where students live.
Collapse
Affiliation(s)
- Avantika R Diwadkar
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, US
| | - Susan Yoon
- Graduate School of Education, University of Pennsylvania, Philadelphia, PA, US
| | - Jooeun Shim
- Graduate School of Education, University of Pennsylvania, Philadelphia, PA, US
| | - Michael Gonzalez
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, US
| | - Ryan Urbanowicz
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, US
| | - Blanca E Himes
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, US
| |
Collapse
|
19
|
Cheung H, Yu J. Machine learning on microbiome research in gastrointestinal cancer. J Gastroenterol Hepatol 2021; 36:817-822. [PMID: 33880761 DOI: 10.1111/jgh.15502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 03/18/2021] [Indexed: 12/24/2022]
Abstract
Gastrointestinal cancer maintains the highest incidence and mortality rate among all cancers globally. In addition to genetic causes, it has been reported that individuals' diet and composition of the gastrointestinal microbiome have profound impacts on gastrointestinal cancer development. Microbiome research has risen in popularity to provide alternative insights into cancer development and potential therapeutic effect. However, there is a lack of an effective analytical tool to comprehend the massive amount of data generated from high-throughput sequencing methods. Artificial intelligence is another rapidly developing field that has strong application potential in microbiome research. Deep learning and machine learning are two subfields under the umbrella of artificial intelligence. Here we discuss the current approaches to study the gut microbiome, as well as the applications and challenges of implementing artificial intelligence in microbiome research.
Collapse
Affiliation(s)
- Henley Cheung
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
20
|
Cammarota G, Ianiro G, Ahern A, Carbone C, Temko A, Claesson MJ, Gasbarrini A, Tortora G. Gut microbiome, big data and machine learning to promote precision medicine for cancer. Nat Rev Gastroenterol Hepatol 2020; 17:635-648. [PMID: 32647386 DOI: 10.1038/s41575-020-0327-3] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/02/2020] [Indexed: 12/13/2022]
Abstract
The gut microbiome has been implicated in cancer in several ways, as specific microbial signatures are known to promote cancer development and influence safety, tolerability and efficacy of therapies. The 'omics' technologies used for microbiome analysis continuously evolve and, although much of the research is still at an early stage, large-scale datasets of ever increasing size and complexity are being produced. However, there are varying levels of difficulty in realizing the full potential of these new tools, which limit our ability to critically analyse much of the available data. In this Perspective, we provide a brief overview on the role of gut microbiome in cancer and focus on the need, role and limitations of a machine learning-driven approach to analyse large amounts of complex health-care information in the era of big data. We also discuss the potential application of microbiome-based big data aimed at promoting precision medicine in cancer.
Collapse
Affiliation(s)
- Giovanni Cammarota
- Gastroenterology Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Gianluca Ianiro
- Gastroenterology Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Ahern
- School of Microbiology and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Carmine Carbone
- Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andriy Temko
- School of Engineering, University College Cork, Cork, Ireland.,Qualcomm ML R&D, Cork, Ireland
| | - Marcus J Claesson
- School of Microbiology and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Antonio Gasbarrini
- Gastroenterology Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giampaolo Tortora
- Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
21
|
Fleischhacker SE, Woteki CE, Coates PM, Hubbard VS, Flaherty GE, Glickman DR, Harkin TR, Kessler D, Li WW, Loscalzo J, Parekh A, Rowe S, Stover PJ, Tagtow A, Yun AJ, Mozaffarian D. Strengthening national nutrition research: rationale and options for a new coordinated federal research effort and authority. Am J Clin Nutr 2020; 112:721-769. [PMID: 32687145 PMCID: PMC7454258 DOI: 10.1093/ajcn/nqaa179] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The US faces remarkable food and nutrition challenges. A new federal effort to strengthen and coordinate nutrition research could rapidly generate the evidence base needed to address these multiple national challenges. However, the relevant characteristics of such an effort have been uncertain. OBJECTIVES Our aim was to provide an objective, informative summary of 1) the mounting diet-related health burdens facing our nation and corresponding economic, health equity, national security, and sustainability implications; 2) the current federal nutrition research landscape and existing mechanisms for its coordination; 3) the opportunities for and potential impact of new fundamental, clinical, public health, food and agricultural, and translational scientific discoveries; and 4) the various options for further strengthening and coordinating federal nutrition research, including corresponding advantages, disadvantages, and potential executive and legislative considerations. METHODS We reviewed government and other published documents on federal nutrition research; held various discussions with expert groups, advocacy organizations, and scientific societies; and held in-person or phone meetings with >50 federal staff in executive and legislative roles, as well as with a variety of other stakeholders in academic, industry, and nongovernment organizations. RESULTS Stark national nutrition challenges were identified. More Americans are sick than are healthy, largely from rising diet-related illnesses. These conditions create tremendous strains on productivity, health care costs, health disparities, government budgets, US economic competitiveness, and military readiness. The coronavirus disease 2019 (COVID-19) outbreak has further laid bare these strains, including food insecurity, major diet-related comorbidities for poor outcomes from COVID-19 such as diabetes, hypertension, and obesity, and insufficient surveillance on and coordination of our food system. More than 10 federal departments and agencies currently invest in critical nutrition research, yet with relatively flat investments over several decades. Coordination also remains suboptimal, documented by multiple governmental reports over 50 years. Greater harmonization and expansion of federal investment in nutrition science, not a silo-ing or rearrangement of existing investments, has tremendous potential to generate new discoveries to improve and sustain the health of all Americans. Two identified key strategies to achieve this were as follows: 1) a new authority for robust cross-governmental coordination of nutrition research and other nutrition-related policy and 2) strengthened authority, investment, and coordination for nutrition research within the NIH. These strategies were found to be complementary, together catalyzing important new science, partnerships, coordination, and returns on investment. Additional complementary actions to accelerate federal nutrition research were identified at the USDA. CONCLUSIONS The need and opportunities for strengthened federal nutrition research are clear, with specific identified options to help create the new leadership, strategic planning, coordination, and investment the nation requires to address the multiple nutrition-related challenges and grasp the opportunities before us.
Collapse
Affiliation(s)
| | - Catherine E Woteki
- University of Virginia Biocomplexity Institute and Initiative, Arlington, VA, USA
| | - Paul M Coates
- Retired, National Institutes of Health, Bethesda, MD, USA
| | - Van S Hubbard
- Retired, National Institutes of Health, Bethesda, MD, USA
| | - Grace E Flaherty
- Gerald J and Dorothy R Friedman School of Nutrition Science and Policy at Tufts University, Boston, MA, USA
| | | | | | - David Kessler
- Former Food and Drug Administration Commissioner, College Park, MD, USA
| | | | - Joseph Loscalzo
- Department of Medicine at Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Patrick J Stover
- Texas A&M AgriLife, Texas A&M College of Agriculture and Life Sciences, and Texas A&M AgriLife Research, College Station, TX, USA
| | | | | | - Dariush Mozaffarian
- Gerald J and Dorothy R Friedman School of Nutrition Science and Policy at Tufts University, Boston, MA, USA
| |
Collapse
|
22
|
Gabryszewski SJ, Chang X, Dudley JW, Mentch F, March M, Holmes JH, Moore J, Grundmeier RW, Hakonarson H, Hill DA. Unsupervised modeling and genome-wide association identify novel features of allergic march trajectories. J Allergy Clin Immunol 2020; 147:677-685.e10. [PMID: 32650023 DOI: 10.1016/j.jaci.2020.06.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/07/2020] [Accepted: 06/05/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND The allergic march refers to the natural history of allergic conditions during infancy and childhood. However, population-level disease incidence patterns do not necessarily reflect the development of allergic disease in individuals. A better understanding of the factors that predispose to different allergic trajectories is needed. OBJECTIVE Our aim was to determine the demographic and genetic features that are associated with the major allergic march trajectories. METHODS Presence or absence of common allergic conditions (atopic dermatitis [AD], IgE-mediated food allergy [IgE-FA], asthma, and allergic rhinitis [AR]) was ascertained in a pediatric primary care birth cohort of 158,510 subjects. Hierarchic clustering and decision tree modeling were used to associate demographic features with allergic outcomes. Genome-wide association study was used to test for risk loci associated with specific allergic trajectories. RESULTS We found an association between self-identified black race and progression from AD to asthma. Conversely, Asian or Pacific Islander race was associated with progression from AD to IgE-mediated food allergy, and white race was associated with progression from AD to AR. Genome-wide association study of trajectory groups identified risk loci associated with progression from AD to asthma (rs60242841) and from AD to AR (rs9565267, rs151041509, and rs78171803). Consistent with our epidemiologic associations, rs60242841 was more common in individuals of African ancestry than in individuals of European ancestry, whereas rs9565267 and rs151041509 were more common in individuals of European ancestry than in individuals of African ancestry. CONCLUSION We have identified novel associations between race and progression along distinct allergic trajectories. Ancestral genetic differences may contribute to these associations. These results uncover important health disparities, refine the concept of the allergic march, and represent a step toward developing individualized medical approaches for these conditions.
Collapse
Affiliation(s)
| | - Xiao Chang
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Jesse W Dudley
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Frank Mentch
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Michael March
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - John H Holmes
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Jason Moore
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Robert W Grundmeier
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pa; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pa; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa; Division of Pulmonary Medicine, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - David A Hill
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pa; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa.
| |
Collapse
|
23
|
Affiliation(s)
- Ebony Torrington
- Future Science Group, Unitec House, 2 Albert Place, London, N3 1QB, UK
| |
Collapse
|