1
|
Serra A, Fratello M, Federico A, Greco D. An update on knowledge graphs and their current and potential applications in drug discovery. Expert Opin Drug Discov 2025; 20:599-619. [PMID: 40223439 DOI: 10.1080/17460441.2025.2490253] [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: 09/19/2024] [Accepted: 04/03/2025] [Indexed: 04/15/2025]
Abstract
INTRODUCTION Knowledge graphs are becoming prominent tools in computational drug discovery. They effectively integrate heterogeneous biomedical data and generate new hypotheses and knowledge. AREAS COVERED This article is based on a literature review using Google Scholar and PubMed to retrieve articles on existing knowledge graphs relevant to the drug discovery field. The authors compare the types of entities, relationships, and data sources they encompass. Additionally, the authors provide examples of their use in the drug discovery field and discuss potential strategies for advancing this research area. EXPERT OPINION Knowledge graphs are crucial in drug discovery, but their construction leads to challenges in data integration and consistency. Future research should prioritize the standardization of data sources and data modeling. More efforts are needed for the integration in knowledge graphs of diverse data types, such as chemical structures and epigenetic data, to enhance their effectiveness. Additionally, advancements in large language models should be pursued to aid the development of knowledge graphs, provide intuitive querying capabilities for non-expert users, and explain knowledge graphs -derived predictions, thereby making these tools more accessible and their insights more interpretable for a wider audience.
Collapse
Affiliation(s)
- Angela Serra
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
| | - Michele Fratello
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Antonio Federico
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
| | - Dario Greco
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
2
|
Freidel S, Schwarz E. Knowledge graphs in psychiatric research: Potential applications and future perspectives. Acta Psychiatr Scand 2025; 151:180-191. [PMID: 38886846 PMCID: PMC11787922 DOI: 10.1111/acps.13717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/15/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Knowledge graphs (KGs) remain an underutilized tool in the field of psychiatric research. In the broader biomedical field KGs are already a significant tool mainly used as knowledge database or for novel relation detection between biomedical entities. This review aims to outline how KGs would further research in the field of psychiatry in the age of Artificial Intelligence (AI) and Large Language Models (LLMs). METHODS We conducted a thorough literature review across a spectrum of scientific fields ranging from computer science and knowledge engineering to bioinformatics. The literature reviewed was taken from PubMed, Semantic Scholar and Google Scholar searches including terms such as "Psychiatric Knowledge Graphs", "Biomedical Knowledge Graphs", "Knowledge Graph Machine Learning Applications", "Knowledge Graph Applications for Biomedical Sciences". The resulting publications were then assessed and accumulated in this review regarding their possible relevance to future psychiatric applications. RESULTS A multitude of papers and applications of KGs in associated research fields that are yet to be utilized in psychiatric research was found and outlined in this review. We create a thorough recommendation for other computational researchers regarding use-cases of these KG applications in psychiatry. CONCLUSION This review illustrates use-cases of KG-based research applications in biomedicine and beyond that may aid in elucidating the complex biology of psychiatric illness and open new routes for developing innovative interventions. We conclude that there is a wealth of opportunities for KG utilization in psychiatric research across a variety of application areas including biomarker discovery, patient stratification and personalized medicine approaches.
Collapse
Affiliation(s)
- Sebastian Freidel
- Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Emanuel Schwarz
- Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| |
Collapse
|
3
|
Prasanna S, Kumar A, Rao D, Simoes EJ, Rao P. A scalable tool for analyzing genomic variants of humans using knowledge graphs and graph machine learning. Front Big Data 2025; 7:1466391. [PMID: 39906190 PMCID: PMC11790625 DOI: 10.3389/fdata.2024.1466391] [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: 07/17/2024] [Accepted: 12/12/2024] [Indexed: 02/06/2025] Open
Abstract
Advances in high-throughput genome sequencing have enabled large-scale genome sequencing in clinical practice and research studies. By analyzing genomic variants of humans, scientists can gain better understanding of the risk factors of complex diseases such as cancer and COVID-19. To model and analyze the rich genomic data, knowledge graphs (KGs) and graph machine learning (GML) can be regarded as enabling technologies. In this article, we present a scalable tool called VariantKG for analyzing genomic variants of humans modeled using KGs and GML. Specifically, we used publicly available genome sequencing data from patients with COVID-19. VariantKG extracts variant-level genetic information output by a variant calling pipeline, annotates the variant data with additional metadata, and converts the annotated variant information into a KG represented using the Resource Description Framework (RDF). The resulting KG is further enhanced with patient metadata and stored in a scalable graph database that enables efficient RDF indexing and query processing. VariantKG employs the Deep Graph Library (DGL) to perform GML tasks such as node classification. A user can extract a subset of the KG and perform inference tasks using DGL. The user can monitor the training and testing performance and hardware utilization. We tested VariantKG for KG construction by using 1,508 genome sequences, leading to 4 billion RDF statements. We evaluated GML tasks using VariantKG by selecting a subset of 500 sequences from the KG and performing node classification using well-known GML techniques such as GraphSAGE, Graph Convolutional Network (GCN) and Graph Transformer. VariantKG has intuitive user interfaces and features enabling a low barrier to entry for KG construction, model inference, and model interpretation on genomic variants of humans.
Collapse
Affiliation(s)
- Shivika Prasanna
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, United States
| | - Ajay Kumar
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, United States
| | - Deepthi Rao
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, United States
| | - Eduardo J. Simoes
- Department of Biomedical Informatics, Biostatistics and Medical Epidemiology, University of Missouri, Columbia, MO, United States
| | - Praveen Rao
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, United States
| |
Collapse
|
4
|
Mohseni Ahooyi T, Stear B, Simmons JA, Nemarich CM, Silverstein JC, Taylor DM. Homo Sapiens Chromosomal Location Ontology: A Framework for Genomic Data in Biomedical Knowledge Graphs. Sci Data 2025; 12:52. [PMID: 39799122 PMCID: PMC11724927 DOI: 10.1038/s41597-024-04358-x] [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: 02/21/2024] [Accepted: 12/20/2024] [Indexed: 01/15/2025] Open
Abstract
The Homo sapiens Chromosomal Location Ontology (HSCLO) is designed to facilitate the integration of human genomic features into biomedical knowledge graphs from releases GRCh37 and GRCh38 at multiple resolutions. HSCLO comprises two distinct versions, HSCLO37 and HSCLO38, each tailored to its respective human genome release. This ontology supports the efficient integration and analysis of human genomic data across scales ranging from entire chromosomes to individual base pairs, thereby enhancing data retrieval and interoperability within large-scale biomedical datasets. Unlike existing ontologies that primarily focus on genomic feature identification or annotation, HSCLO is specifically engineered to optimize the interoperability and scalability of genomic data within biomedical knowledge graphs. The utility and performance of HSCLO are demonstrated through a case study involving the integration of high-resolution chromatin interaction data, which reveals significant improvements in query efficiency and data linkage. HSCLO represents a valuable resource for advancing research in disease genetics, personalized medicine, and other domains that require complex genomic data integration.
Collapse
Affiliation(s)
- Taha Mohseni Ahooyi
- The Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Benjamin Stear
- The Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - J Alan Simmons
- Department of Biomedical Informatics, School of Medicine, The University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher M Nemarich
- The Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan C Silverstein
- Department of Biomedical Informatics, School of Medicine, The University of Pittsburgh, Pittsburgh, PA, USA
| | - Deanne M Taylor
- The Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, University of Pennsylvania Perelman Medical School, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Stear BJ, Mohseni Ahooyi T, Simmons JA, Kollar C, Hartman L, Beigel K, Lahiri A, Vasisht S, Callahan TJ, Nemarich CM, Silverstein JC, Taylor DM. Petagraph: A large-scale unifying knowledge graph framework for integrating biomolecular and biomedical data. Sci Data 2024; 11:1338. [PMID: 39695169 PMCID: PMC11655564 DOI: 10.1038/s41597-024-04070-w] [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: 05/24/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
Over the past decade, there has been substantial growth in both the quantity and complexity of available biomedical data. In order to more efficiently harness this extensive data and alleviate challenges associated with integration of multi-omics data, we developed Petagraph, a biomedical knowledge graph that encompasses over 32 million nodes and 118 million relationships. Petagraph leverages more than 180 ontologies and standards in the Unified Biomedical Knowledge Graph (UBKG) to embed millions of quantitative genomics data points. Petagraph provides a cohesive data environment that enables users to efficiently analyze, annotate, and discern relationships within and across complex multi-omics datasets supported by UBKG's annotation scaffold. We demonstrate how queries on Petagraph can generate meaningful results across various research contexts and use cases.
Collapse
Affiliation(s)
- Benjamin J Stear
- Department of Biomedical and Health Informatics (DBHI), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Taha Mohseni Ahooyi
- Department of Biomedical and Health Informatics (DBHI), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - J Alan Simmons
- Department of Biomedical Informatics, School of Medicine, The University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles Kollar
- Department of Biomedical Informatics, School of Medicine, The University of Pittsburgh, Pittsburgh, PA, USA
| | - Lance Hartman
- Department of Biomedical and Health Informatics (DBHI), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Katherine Beigel
- Department of Biomedical and Health Informatics (DBHI), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Aditya Lahiri
- Department of Biomedical and Health Informatics (DBHI), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shubha Vasisht
- Department of Biomedical and Health Informatics (DBHI), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tiffany J Callahan
- Department of Biomedical Informatics, Columbia University Irving Medical Campus, New York, NY, USA
| | - Christopher M Nemarich
- Department of Biomedical and Health Informatics (DBHI), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan C Silverstein
- Department of Biomedical Informatics, School of Medicine, The University of Pittsburgh, Pittsburgh, PA, USA
| | - Deanne M Taylor
- Department of Biomedical and Health Informatics (DBHI), The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, University of Pennsylvania Perelman Medical School, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Johnson R, Gottlieb U, Shaham G, Eisen L, Waxman J, Devons-Sberro S, Ginder CR, Hong P, Sayeed R, Reis BY, Balicer RD, Dagan N, Zitnik M. Unified Clinical Vocabulary Embeddings for Advancing Precision Medicine. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.03.24318322. [PMID: 39677476 PMCID: PMC11643188 DOI: 10.1101/2024.12.03.24318322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Integrating clinical knowledge into AI remains challenging despite numerous medical guidelines and vocabularies. Medical codes, central to healthcare systems, often reflect operational patterns shaped by geographic factors, national policies, insurance frameworks, and physician practices rather than the precise representation of clinical knowledge. This disconnect hampers AI in representing clinical relationships, raising concerns about bias, transparency, and generalizability. Here, we developed a resource of 67,124 clinical vocabulary embeddings derived from a clinical knowledge graph tailored to electronic health record vocabularies, spanning over 1.3 million edges. Using graph transformer neural networks, we generated clinical vocabulary embeddings that provide a new representation of clinical knowledge by unifying seven medical vocabularies. These embeddings were validated through a phenotype risk score analysis involving 4.57 million patients from Clalit Healthcare Services, effectively stratifying individuals based on survival outcomes. Inter-institutional panels of clinicians evaluated the embeddings for alignment with clinical knowledge across 90 diseases and 3,000 clinical codes, confirming their robustness and transferability. This resource addresses gaps in integrating clinical vocabularies into AI models and training datasets, paving the way for knowledge-grounded population and patient-level models.
Collapse
Affiliation(s)
- Ruth Johnson
- The Ivan and Francesca Berkowitz Family Living Laboratory Collaboration at Harvard Medical School and Clalit Research Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Uri Gottlieb
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
| | - Galit Shaham
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
| | - Lihi Eisen
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
| | - Jacob Waxman
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
| | - Stav Devons-Sberro
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
| | - Curtis R. Ginder
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter Hong
- Division of General Pediatrics, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Information Technology, Enterprise Data Analytics and Reporting, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Raheel Sayeed
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Ben Y. Reis
- The Ivan and Francesca Berkowitz Family Living Laboratory Collaboration at Harvard Medical School and Clalit Research Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Predictive Medicine Group, Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, USA
- Harvard Data Science Initiative, Cambridge, MA, USA
| | - Ran D. Balicer
- The Ivan and Francesca Berkowitz Family Living Laboratory Collaboration at Harvard Medical School and Clalit Research Institute, Boston, MA, USA
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
- Faculty of Health Sciences, School of Public Health, Ben Gurion University of the Negev, Be’er Sheva, Israel
| | - Noa Dagan
- The Ivan and Francesca Berkowitz Family Living Laboratory Collaboration at Harvard Medical School and Clalit Research Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
- Software and Information Systems Engineering, Ben Gurion University, Be’er Sheva, Israel
| | - Marinka Zitnik
- The Ivan and Francesca Berkowitz Family Living Laboratory Collaboration at Harvard Medical School and Clalit Research Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Harvard Data Science Initiative, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Allston, MA, USA
| |
Collapse
|
7
|
Ceskoutsé RFT, Bomgni AB, Gnimpieba Zanfack DR, Agany DDM, Thomas BB, Zohim EG. HeteroKGRep: Heterogeneous Knowledge Graph based Drug Repositioning. Knowl Based Syst 2024; 305:112638. [PMID: 39610660 PMCID: PMC11600970 DOI: 10.1016/j.knosys.2024.112638] [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] [Indexed: 11/30/2024]
Abstract
The process of developing new drugs is both time-consuming and costly, often taking over a decade and billions of dollars to obtain regulatory approval. Additionally, the complexity of patent protection for novel compounds presents challenges for pharmaceutical innovation. Drug repositioning offers an alternative strategy to uncover new therapeutic uses for existing medicines. Previous repositioning models have been limited by their reliance on homogeneous data sources, failing to leverage the rich information available in heterogeneous biomedical knowledge graphs. We propose HeteroKGRep, a novel drug repositioning model that utilizes heterogeneous graphs to address these limitations. HeteroKGRep is a multi-step framework that first generates a similarity graph from hierarchical concept relations. It then applies SMOTE over-sampling to address class imbalance before generating node sequences using a heterogeneous graph neural network. Drug and disease embeddings are extracted from the network and used for prediction. We evaluated HeteroKGRep on a graph containing biomedical concepts and relations from ontologies, pathways and literature. It achieved state-of-the-art performance with 99% accuracy, 95% AUC ROC and 94% average precision on predicting repurposing opportunities. Compared to existing homogeneous approaches, HeteroKGRep leverages diverse knowledge sources to enrich representation learning. Based on heterogeneous graphs, HeteroKGRep can discover new drug-desease associations, leveraging de novo drug development. This work establishes a promising new paradigm for knowledge-guided drug repositioning using multimodal biomedical data.
Collapse
Affiliation(s)
- Ribot Fleury T Ceskoutsé
- Ecole Nationale Supérieure Polytechnique, University of Yaounde I, P.O. Box. 8390, Yaoundé, Cameroon
| | - Alain Bertrand Bomgni
- University of South Dakota, 4800 N Career Avenue, 57107, SD, USA
- Departement of Mathematics and computer science, University of Dschang, P.O. Box. 67, Dschang, Cameroon
| | - David R Gnimpieba Zanfack
- Laboratory of Innovative Technologies (LTI), University of Picardie Jule Verne (UPJV), 48 Rue Raspail, 02100 Saint Quentin, France
| | - Diing D M Agany
- University of South Dakota, 4800 N Career Avenue, 57107, SD, USA
| | - Bouetou Bouetou Thomas
- Ecole Nationale Supérieure Polytechnique, University of Yaounde I, P.O. Box. 8390, Yaoundé, Cameroon
| | | |
Collapse
|
8
|
Man J, Shi Y, Hu Z, Yang R, Huang Z, Zhou Y. KSDKG: construction and application of knowledge graph for kidney stone disease based on biomedical literature and public databases. Health Inf Sci Syst 2024; 12:54. [PMID: 39554224 PMCID: PMC11564440 DOI: 10.1007/s13755-024-00309-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 09/22/2024] [Indexed: 11/19/2024] Open
Abstract
Purpose Kidney stone disease (KSD) is a common urological disorder with an increasing incidence worldwide. The extensive knowledge about KSD is dispersed across multiple databases, challenging the visualization and representation of its hierarchy and connections. This paper aims at constructing a disease-specific knowledge graph for KSD to enhance the effective utilization of knowledge by medical professionals and promote clinical research and discovery. Methods Text parsing and semantic analysis were conducted on literature related to KSD from PubMed, with concept annotation based on biomedical ontology being utilized to generate semantic data in RDF format. Moreover, public databases were integrated to construct a large-scale knowledge graph for KSD. Additionally, case studies were carried out to demonstrate the practical utility of the developed knowledge graph. Results We proposed and implemented a Kidney Stone Disease Knowledge Graph (KSDKG), covering more than 90 million triples. This graph comprised semantic data extracted from 29,174 articles, integrating available data from UMLS, SNOMED CT, MeSH, DrugBank and Microbe-Disease Knowledge Graph. Through the application of three cases, we retrieved and discovered information on microbes, drugs and diseases associated with KSD. The results illustrated that the KSDKG can integrate diverse medical knowledge and provide new clinical insights for identifying the underlying mechanisms of KSD. Conclusion The KSDKG efficiently utilizes knowledge graph to reveal hidden knowledge associations, facilitating semantic search and response. As a blueprint for developing disease-specific knowledge graphs, it offers valuable contributions to medical research.
Collapse
Affiliation(s)
- Jianping Man
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Yufei Shi
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Zhensheng Hu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Rui Yang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Zhisheng Huang
- Knowledge Representation and Reasoning (KR &R) Group, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yi Zhou
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080 China
| |
Collapse
|
9
|
Deciphering the impact of genomic variation on function. Nature 2024; 633:47-57. [PMID: 39232149 PMCID: PMC11973978 DOI: 10.1038/s41586-024-07510-0] [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: 04/11/2023] [Accepted: 05/02/2024] [Indexed: 09/06/2024]
Abstract
Our genomes influence nearly every aspect of human biology-from molecular and cellular functions to phenotypes in health and disease. Studying the differences in DNA sequence between individuals (genomic variation) could reveal previously unknown mechanisms of human biology, uncover the basis of genetic predispositions to diseases, and guide the development of new diagnostic tools and therapeutic agents. Yet, understanding how genomic variation alters genome function to influence phenotype has proved challenging. To unlock these insights, we need a systematic and comprehensive catalogue of genome function and the molecular and cellular effects of genomic variants. Towards this goal, the Impact of Genomic Variation on Function (IGVF) Consortium will combine approaches in single-cell mapping, genomic perturbations and predictive modelling to investigate the relationships among genomic variation, genome function and phenotypes. IGVF will create maps across hundreds of cell types and states describing how coding variants alter protein activity, how noncoding variants change the regulation of gene expression, and how such effects connect through gene-regulatory and protein-interaction networks. These experimental data, computational predictions and accompanying standards and pipelines will be integrated into an open resource that will catalyse community efforts to explore how our genomes influence biology and disease across populations.
Collapse
|
10
|
Mulero-Hernández J, Mironov V, Miñarro-Giménez JA, Kuiper M, Fernández-Breis J. Integration of chromosome locations and functional aspects of enhancers and topologically associating domains in knowledge graphs enables versatile queries about gene regulation. Nucleic Acids Res 2024; 52:e69. [PMID: 38967009 PMCID: PMC11347148 DOI: 10.1093/nar/gkae566] [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: 01/12/2023] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024] Open
Abstract
Knowledge about transcription factor binding and regulation, target genes, cis-regulatory modules and topologically associating domains is not only defined by functional associations like biological processes or diseases but also has a determinative genome location aspect. Here, we exploit these location and functional aspects together to develop new strategies to enable advanced data querying. Many databases have been developed to provide information about enhancers, but a schema that allows the standardized representation of data, securing interoperability between resources, has been lacking. In this work, we use knowledge graphs for the standardized representation of enhancers and topologically associating domains, together with data about their target genes, transcription factors, location on the human genome, and functional data about diseases and gene ontology annotations. We used this schema to integrate twenty-five enhancer datasets and two domain datasets, creating the most powerful integrative resource in this field to date. The knowledge graphs have been implemented using the Resource Description Framework and integrated within the open-access BioGateway knowledge network, generating a resource that contains an interoperable set of knowledge graphs (enhancers, TADs, genes, proteins, diseases, GO terms, and interactions between domains). We show how advanced queries, which combine functional and location restrictions, can be used to develop new hypotheses about functional aspects of gene expression regulation.
Collapse
Affiliation(s)
- Juan Mulero-Hernández
- Departamento de Informática y Sistemas, Universidad de Murcia, CEIR Campus Mare Nostrum, Instituto Murciano de Investigación Biosanitaria (IMIB),30100 Murcia, Spain
| | - Vladimir Mironov
- Department of Biology, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - José Antonio Miñarro-Giménez
- Departamento de Informática y Sistemas, Universidad de Murcia, CEIR Campus Mare Nostrum, Instituto Murciano de Investigación Biosanitaria (IMIB),30100 Murcia, Spain
| | - Martin Kuiper
- Department of Biology, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Jesualdo Tomás Fernández-Breis
- Departamento de Informática y Sistemas, Universidad de Murcia, CEIR Campus Mare Nostrum, Instituto Murciano de Investigación Biosanitaria (IMIB),30100 Murcia, Spain
| |
Collapse
|
11
|
Middleton L, Melas I, Vasavda C, Raies A, Rozemberczki B, Dhindsa RS, Dhindsa JS, Weido B, Wang Q, Harper AR, Edwards G, Petrovski S, Vitsios D. Phenome-wide identification of therapeutic genetic targets, leveraging knowledge graphs, graph neural networks, and UK Biobank data. SCIENCE ADVANCES 2024; 10:eadj1424. [PMID: 38718126 PMCID: PMC11078195 DOI: 10.1126/sciadv.adj1424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 04/04/2024] [Indexed: 05/12/2024]
Abstract
The ongoing expansion of human genomic datasets propels therapeutic target identification; however, extracting gene-disease associations from gene annotations remains challenging. Here, we introduce Mantis-ML 2.0, a framework integrating AstraZeneca's Biological Insights Knowledge Graph and numerous tabular datasets, to assess gene-disease probabilities throughout the phenome. We use graph neural networks, capturing the graph's holistic structure, and train them on hundreds of balanced datasets via a robust semi-supervised learning framework to provide gene-disease probabilities across the human exome. Mantis-ML 2.0 incorporates natural language processing to automate disease-relevant feature selection for thousands of diseases. The enhanced models demonstrate a 6.9% average classification power boost, achieving a median receiver operating characteristic (ROC) area under curve (AUC) score of 0.90 across 5220 diseases from Human Phenotype Ontology, OpenTargets, and Genomics England. Notably, Mantis-ML 2.0 prioritizes associations from an independent UK Biobank phenome-wide association study (PheWAS), providing a stronger form of triaging and mitigating against underpowered PheWAS associations. Results are exposed through an interactive web resource.
Collapse
Affiliation(s)
- Lawrence Middleton
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ioannis Melas
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Chirag Vasavda
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA 02451, USA
| | - Arwa Raies
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Benedek Rozemberczki
- Biological Insights Knowledge Graph (BIKG), Research D&A, R&D IT, AstraZeneca, Cambridge, UK
| | - Ryan S. Dhindsa
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA 02451, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Justin S. Dhindsa
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Blake Weido
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Quanli Wang
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA 02451, USA
| | - Andrew R. Harper
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Gavin Edwards
- Biological Insights Knowledge Graph (BIKG), Research D&A, R&D IT, AstraZeneca, Cambridge, UK
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Department of Medicine, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Dimitrios Vitsios
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
12
|
Ma C, Liu S, Koslicki D. MetagenomicKG: a knowledge graph for metagenomic applications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.14.585056. [PMID: 38559251 PMCID: PMC10980061 DOI: 10.1101/2024.03.14.585056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Motivation The sheer volume and variety of genomic content within microbial communities makes metagenomics a field rich in biomedical knowledge. To traverse these complex communities and their vast unknowns, metagenomic studies often depend on distinct reference databases, such as the Genome Taxonomy Database (GTDB), the Kyoto Encyclopedia of Genes and Genomes (KEGG), and the Bacterial and Viral Bioinformatics Resource Center (BV-BRC), for various analytical purposes. These databases are crucial for genetic and functional annotation of microbial communities. Nevertheless, the inconsistent nomenclature or identifiers of these databases present challenges for effective integration, representation, and utilization. Knowledge graphs (KGs) offer an appropriate solution by organizing biological entities and their interrelations into a cohesive network. The graph structure not only facilitates the unveiling of hidden patterns but also enriches our biological understanding with deeper insights. Despite KGs having shown potential in various biomedical fields, their application in metagenomics remains underexplored. Results We present MetagenomicKG, a novel knowledge graph specifically tailored for metagenomic analysis. MetagenomicKG integrates taxonomic, functional, and pathogenesis-related information from widely used databases, and further links these with established biomedical knowledge graphs to expand biological connections. Through several use cases, we demonstrate its utility in enabling hypothesis generation regarding the relationships between microbes and diseases, generating sample-specific graph embeddings, and providing robust pathogen prediction. Availability and Implementation The source code and technical details for constructing the MetagenomicKG and reproducing all analyses are available at Github: https://github.com/KoslickiLab/MetagenomicKG. We also host a Neo4j instance: http://mkg.cse.psu.edu:7474 for accessing and querying this graph.
Collapse
Affiliation(s)
- Chunyu Ma
- Huck Institutes of the Life Sciences, Pennsylvania State University, State College, Pennsylvania, USA
| | - Shaopeng Liu
- Huck Institutes of the Life Sciences, Pennsylvania State University, State College, Pennsylvania, USA
| | - David Koslicki
- Huck Institutes of the Life Sciences, Pennsylvania State University, State College, Pennsylvania, USA
- Department of Computer Science and Engineering, Pennsylvania State University, State College, Pennsylvania, USA
- Department of Biology, Pennsylvania State University, State College, Pennsylvania, USA
- The One Health Microbiome Center, Huck Institutes of the Life Sciences, Pennsylvania State University, State College, Pennsylvania, USA
| |
Collapse
|
13
|
Jin S, Liang H, Zhang W, Li H. Knowledge Graph for Breast Cancer Prevention and Treatment: Literature-Based Data Analysis Study. JMIR Med Inform 2024; 12:e52210. [PMID: 38409769 PMCID: PMC11004512 DOI: 10.2196/52210] [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: 08/26/2023] [Revised: 01/02/2024] [Accepted: 01/06/2024] [Indexed: 02/28/2024] Open
Abstract
Background The incidence of breast cancer has remained high and continues to rise since the 21st century. Consequently, there has been a significant increase in research efforts focused on breast cancer prevention and treatment. Despite the extensive body of literature available on this subject, systematic integration is lacking. To address this issue, knowledge graphs have emerged as a valuable tool. By harnessing their powerful knowledge integration capabilities, knowledge graphs offer a comprehensive and structured approach to understanding breast cancer prevention and treatment. Objective We aim to integrate literature data on breast cancer treatment and prevention, build a knowledge graph, and provide support for clinical decision-making. Methods We used Medical Subject Headings terms to search for clinical trial literature on breast cancer prevention and treatment published on PubMed between 2018 and 2022. We downloaded triplet data from the Semantic MEDLINE Database (SemMedDB) and matched them with the retrieved literature to obtain triplet data for the target articles. We visualized the triplet information using NetworkX for knowledge discovery. Results Within the scope of literature research in the past 5 years, malignant neoplasms appeared most frequently (587/1387, 42.3%). Pharmacotherapy (267/1387, 19.3%) was the primary treatment method, with trastuzumab (209/1805, 11.6%) being the most commonly used therapeutic drug. Through the analysis of the knowledge graph, we have discovered a complex network of relationships between treatment methods, therapeutic drugs, and preventive measures for different types of breast cancer. Conclusions This study constructed a knowledge graph for breast cancer prevention and treatment, which enabled the integration and knowledge discovery of relevant literature in the past 5 years. Researchers can gain insights into treatment methods, drugs, preventive knowledge regarding adverse reactions to treatment, and the associations between different knowledge domains from the graph.
Collapse
Affiliation(s)
- Shuyan Jin
- Health Department, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Haobin Liang
- School of Economics and Statistics, Guangzhou University, Guangzhou, China
| | - Wenxia Zhang
- Health Department, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Huan Li
- Health Department, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, China
| |
Collapse
|
14
|
Park H, Son J, Min J, Choi J. Selective UMLS knowledge infusion for biomedical question answering. Sci Rep 2023; 13:14214. [PMID: 37648800 PMCID: PMC10468517 DOI: 10.1038/s41598-023-41423-8] [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: 02/03/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023] Open
Abstract
One of the artificial intelligence applications in the biomedical field is knowledge-intensive question-answering. As domain expertise is particularly crucial in this field, we propose a method for efficiently infusing biomedical knowledge into pretrained language models, ultimately targeting biomedical question-answering. Transferring all semantics of a large knowledge graph into the entire model requires too many parameters, increasing computational cost and time. We investigate an efficient approach that leverages adapters to inject Unified Medical Language System knowledge into pretrained language models, and we question the need to use all semantics in the knowledge graph. This study focuses on strategies of partitioning knowledge graph and either discarding or merging some for more efficient pretraining. According to the results of three biomedical question answering finetuning datasets, the adapters pretrained on semantically partitioned group showed more efficient performance in terms of evaluation metrics, required parameters, and time. The results also show that discarding groups with fewer concepts is a better direction for small datasets, and merging these groups is better for large dataset. Furthermore, the metric results show a slight improvement, demonstrating that the adapter methodology is rather insensitive to the group formulation.
Collapse
Affiliation(s)
- Hyeryun Park
- Interdisciplinary Program for Bioengineering, Seoul National University Graduate School, Seoul, Republic of Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Jiye Son
- Interdisciplinary Program for Bioengineering, Seoul National University Graduate School, Seoul, Republic of Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Jeongwon Min
- Interdisciplinary Program for Bioengineering, Seoul National University Graduate School, Seoul, Republic of Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Jinwook Choi
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Republic of Korea.
- Department of Biomedical Engineering, College of Medicine, Seoul National University, 103, Daehak-ro, Jongno-gu, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Negro A, Montagna F, Teng MN, Neal T, Thomas S, King S, Khan R. Analysis of the evolution of COVID-19 disease understanding through temporal knowledge graphs. Front Res Metr Anal 2023; 8:1204801. [PMID: 37601534 PMCID: PMC10435856 DOI: 10.3389/frma.2023.1204801] [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: 04/12/2023] [Accepted: 07/06/2023] [Indexed: 08/22/2023] Open
Abstract
The COVID-19 pandemic highlighted two critical barriers hindering rapid response to novel pathogens. These include inefficient use of existing biological knowledge about treatments, compounds, gene interactions, proteins, etc. to fight new diseases, and the lack of assimilation and analysis of the fast-growing knowledge about new diseases to quickly develop new treatments, vaccines, and compounds. Overcoming these critical challenges has the potential to revolutionize global preparedness for future pandemics. Accordingly, this article introduces a novel knowledge graph application that functions as both a repository of life science knowledge and an analytics platform capable of extracting time-sensitive insights to uncover evolving disease dynamics and, importantly, researchers' evolving understanding. Specifically, we demonstrate how to extract time-bounded key concepts, also leveraging existing ontologies, from evolving scholarly articles to create a single temporal connected source of truth specifically related to COVID-19. By doing so, current knowledge can be promptly accessed by both humans and machines, from which further understanding of disease outbreaks can be derived. We present key findings from the temporal analysis, applied to a subset of the resulting knowledge graph known as the temporal keywords knowledge graph, and delve into the detailed capabilities provided by this innovative approach.
Collapse
Affiliation(s)
| | | | - Michael N. Teng
- Division of Allergy and Immunology, Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Tempestt Neal
- Cyber Identity and Behavior Research Laboratory, Department of Computer Science and Engineering, College of Engineering, University of South Florida, Tampa, FL, United States
| | - Sylvia Thomas
- Advanced Membrane and Materials Bio and Integration Research Laboratory, Department of Electrical Engineering, College of Engineering, University of South Florida, Tampa, FL, United States
| | - Sayde King
- Cyber Identity and Behavior Research Laboratory, Department of Computer Science and Engineering, College of Engineering, University of South Florida, Tampa, FL, United States
| | - Ridita Khan
- Advanced Membrane and Materials Bio and Integration Research Laboratory, Department of Electrical Engineering, College of Engineering, University of South Florida, Tampa, FL, United States
| |
Collapse
|
16
|
The Impact of Genomic Variation on Function (IGVF) Consortium. ARXIV 2023:arXiv:2307.13708v1. [PMID: 37547663 PMCID: PMC10402186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Our genomes influence nearly every aspect of human biology from molecular and cellular functions to phenotypes in health and disease. Human genetics studies have now associated hundreds of thousands of differences in our DNA sequence ("genomic variation") with disease risk and other phenotypes, many of which could reveal novel mechanisms of human biology and uncover the basis of genetic predispositions to diseases, thereby guiding the development of new diagnostics and therapeutics. Yet, understanding how genomic variation alters genome function to influence phenotype has proven challenging. To unlock these insights, we need a systematic and comprehensive catalog of genome function and the molecular and cellular effects of genomic variants. Toward this goal, the Impact of Genomic Variation on Function (IGVF) Consortium will combine approaches in single-cell mapping, genomic perturbations, and predictive modeling to investigate the relationships among genomic variation, genome function, and phenotypes. Through systematic comparisons and benchmarking of experimental and computational methods, we aim to create maps across hundreds of cell types and states describing how coding variants alter protein activity, how noncoding variants change the regulation of gene expression, and how both coding and noncoding variants may connect through gene regulatory and protein interaction networks. These experimental data, computational predictions, and accompanying standards and pipelines will be integrated into an open resource that will catalyze community efforts to explore genome function and the impact of genetic variation on human biology and disease across populations.
Collapse
|
17
|
Yang W, Zhao X, Duan L, Niu L, Zhang Y, Zhou W, Li Y, Chen J, Fan A, Xie Q, Liu J, Han Y, Fan D, Hong L. Development and validation of a ligand-receptor pairs signature to predict outcome and provide a therapeutic strategy in gastric cancer. Expert Rev Mol Diagn 2023; 23:619-634. [PMID: 37248704 DOI: 10.1080/14737159.2023.2219843] [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: 12/29/2022] [Accepted: 05/26/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND An important factor in tumor development and progression is the tumor microenvironment (TME), which is heterogeneous. Previous studies have mainly investigated the expression profile and prognostic values of genes in gastric cancer (GC) at the cell population level but neglected the interactions and heterogeneity between cells. METHODS The pattern of ligand-receptor (LR) interactions was delineated on a scRNA-seq dataset containing 44,953 cells from nine GC patients and a fourth bulk RNA-seq dataset including data from 1159 GC patients. We then constructed an LR.Score scoring model to comprehensively evaluate the influence of LR-pairs on the TME, overall survival, and immunotherapy response in GC patients from several cohorts. RESULTS Cell communication network among 13 cell types was constructed based on the LR-pairs. We proposed a new molecular subtyping model for GC based on the LR-pairs and revealed the differences in prognosis, pathophysiologic features, mutation characteristics, function enrichment, and immunological characteristics among the three subtypes. Finally, an LR.Score model based on LR-pairs was developed and validated on several datasets. CONCLUSIONS Based on the selected LR-pairs, we successfully constructed a novel prediction model and observed its well performance on molecular subtyping, target and pathway screening, prognosis judging, and immunotherapy response predicting.
Collapse
Affiliation(s)
- Wanli Yang
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xinhui Zhao
- Department of Thyroid and Breast Surgery, The Affiliated Hospital of Northwest University & Xi'an No.3 Hospital, Northwest University, Xi'an, Shaanxi Province, China
| | - Lili Duan
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Liaoran Niu
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yujie Zhang
- Department of Histology and Embryology, School of Basic Medicine, Xi'an Medical University, Xi'an, Shaanxi Province, China
| | - Wei Zhou
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yiding Li
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Junfeng Chen
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Aqiang Fan
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Qibin Xie
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Jinqiang Liu
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yu Han
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Liu Hong
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|