1
|
Kumar S, Ranga A. Role of miRNAs in breast cancer development and progression: Current research. Biofactors 2025; 51:e2146. [PMID: 39601401 DOI: 10.1002/biof.2146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/16/2024] [Indexed: 11/29/2024]
Abstract
Breast cancer, a complex and heterogeneous ailment impacting numerous women worldwide, persists as a prominent cause of cancer-related fatalities. MicroRNAs (miRNAs), small non-coding RNAs, have garnered significant attention for their involvement in breast cancer's progression. These molecules post-transcriptionally regulate gene expression, influencing crucial cellular processes including proliferation, differentiation, and apoptosis. This review provides an overview of the current research on the role of miRNAs in breast cancer. It discusses the role of miRNAs in breast cancer, including the different subtypes of breast cancer, their molecular characteristics, and the mechanisms by which miRNAs regulate gene expression in breast cancer cells. Additionally, the review highlights recent studies identifying specific miRNAs that are dysregulated in breast cancer and their potential use as diagnostic and prognostic biomarkers. Furthermore, the review explores the therapeutic potential of miRNAs in breast cancer treatment. Preclinical studies have shown the effectiveness of miRNA-based therapies, such as antagomir and miRNA mimic therapies, in inhibiting tumor growth and metastasis. Emerging areas, including the application of artificial intelligence (AI) to advance miRNA research and the "One Health" approach that integrates human and animal cancer insights, are also discussed. However, challenges remain before these therapies can be fully translated into clinical practice. In conclusion, this review emphasizes the significance of miRNAs in breast cancer research and their potential as innovative diagnostic and therapeutic tools. A deeper understanding of miRNA dysregulation in breast cancer is essential for their successful application in clinical settings. With continued research, miRNA-based approaches hold promise for improving patient outcomes in this devastating disease.
Collapse
Affiliation(s)
- Sachin Kumar
- Department of Pharmacology, DIPSAR, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Abhishek Ranga
- Department of Pharmacology, DIPSAR, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| |
Collapse
|
2
|
Sisó S, Kavirayani AM, Couto S, Stierstorfer B, Mohanan S, Morel C, Marella M, Bangari DS, Clark E, Schwartz A, Carreira V. Trends and Challenges of the Modern Pathology Laboratory for Biopharmaceutical Research Excellence. Toxicol Pathol 2024:1926233241303898. [PMID: 39673215 DOI: 10.1177/01926233241303898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2024]
Abstract
Pathology, a fundamental discipline that bridges basic scientific discovery to the clinic, is integral to successful drug development. Intrinsically multimodal and multidimensional, anatomic pathology continues to be empowered by advancements in molecular and digital technologies enabling the spatial tissue detection of biomolecules such as genes, transcripts, and proteins. Over the past two decades, breakthroughs in spatial molecular biology technologies and advancements in automation and digitization of laboratory processes have enabled the implementation of higher throughput assays and the generation of extensive molecular data sets from tissue sections in biopharmaceutical research and development research units. It is our goal to provide readers with some rationale, advice, and ideas to help establish a modern molecular pathology laboratory to meet the emerging needs of biopharmaceutical research. This manuscript provides (1) a high-level overview of the current state and future vision for excellence in research pathology practice and (2) shared perspectives on how to optimally leverage the expertise of discovery, toxicologic, and translational pathologists to provide effective spatial, molecular, and digital pathology data to support modern drug discovery. It captures insights from the experiences, challenges, and solutions from pathology laboratories of various biopharmaceutical organizations, including their approaches to troubleshooting and adopting new technologies.
Collapse
Affiliation(s)
- Sílvia Sisó
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | | | | | | | | | | | - Mathiew Marella
- Janssen Research & Development, LLC, La Jolla, California, USA
| | | | - Elizabeth Clark
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | | | | |
Collapse
|
3
|
Liu Z, Park T. DMOIT: denoised multi-omics integration approach based on transformer multi-head self-attention mechanism. Front Genet 2024; 15:1488683. [PMID: 39720180 PMCID: PMC11666520 DOI: 10.3389/fgene.2024.1488683] [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: 08/30/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Multi-omics data integration has become increasingly crucial for a deeper understanding of the complexity of biological systems. However, effectively integrating and analyzing multi-omics data remains challenging due to their heterogeneity and high dimensionality. Existing methods often struggle with noise, redundant features, and the complex interactions between different omics layers, leading to suboptimal performance. Additionally, they face difficulties in adequately capturing intra-omics interactions due to simplistic concatenation techiniques, and they risk losing critical inter-omics interaction information when using hierarchical attention layers. To address these challenges, we propose a novel Denoised Multi-Omics Integration approach that leverages the Transformer multi-head self-attention mechanism (DMOIT). DMOIT consists of three key modules: a generative adversarial imputation network for handling missing values, a sampling-based robust feature selection module to reduce noise and redundant features, and a multi-head self-attention (MHSA) based feature extractor with a noval architecture that enchance the intra-omics interaction capture. We validated model porformance using cancer datasets from the Cancer Genome Atlas (TCGA), conducting two tasks: survival time classification across different cancer types and estrogen receptor status classification for breast cancer. Our results show that DMOIT outperforms traditional machine learning methods and the state-of-the-art integration method MoGCN in terms of accuracy and weighted F1 score. Furthermore, we compared DMOIT with various alternative MHSA-based architectures to further validate our approach. Our results show that DMOIT consistently outperforms these models across various cancer types and different omics combinations. The strong performance and robustness of DMOIT demonstrate its potential as a valuable tool for integrating multi-omics data across various applications.
Collapse
Affiliation(s)
- Zhe Liu
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
| | - Taesung Park
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
- Department of Statistics, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
4
|
Qin S, Zhang H, Liu C, Yi M. Editorial: Investigating tumor immunotherapy responses in lung cancer using deep learning. Front Immunol 2024; 15:1529949. [PMID: 39691722 PMCID: PMC11649539 DOI: 10.3389/fimmu.2024.1529949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Affiliation(s)
- Shuang Qin
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoxiang Zhang
- Department of Hepatopancreatobiliary Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Chao Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ming Yi
- Department of Breast Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
5
|
Magné N, Sotton S, Varges Gomes A, Marta GN, Giglio RE, Mesía R, Psyrri A, Sacco AG, Shah J, Diao P, Malekzadeh Moghani M, Moreno-Acosta P, Bouleftour W, Deutsch E. Sister partnership to overcome the global burden of cancer. Br J Radiol 2024; 97:1891-1897. [PMID: 39236250 DOI: 10.1093/bjr/tqae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/16/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024] Open
Abstract
Emerging countries are currently facing an increasing burden of cancer while they do not have adequate prevention, monitoring, and research capabilities to tackle the disease. Cancer outcomes are influenced by several factors, including different cancer patterns, national cancer screening guidelines, current stage of disease, and access to quality care and treatments. Discrepancies in cancer care between emerging and developed countries require actions to achieve global health equity. The process of pioneering a sister relationship in the oncology field can thwart the global burden of cancer. The objective of such cooperation programs should include research and training programs, evidence-based oncology practice, and quality cancer. Building global connections will therefore be the novel approach to addressing the global burden of cancer.
Collapse
Affiliation(s)
- Nicolas Magné
- Department of Radiation Oncology, Institut Bergonié, 33076 Bordeaux, France
- Cellular and Molecular Radiobiology Laboratory, Lyon-Sud Medical School, Unité Mixte de Recherche CNRS5822/IP2I, University of Lyon, Oullins 69921, France
| | - Sandrine Sotton
- Medical Oncology Department, Private Loire Hospital (HPL), Saint-Etienne, France
| | - Ana Varges Gomes
- Centro Hospitalar Universitario do Algarve, 8000-386 Faro, Portugal
| | - Gustavo Nader Marta
- Department of Radiation Oncology, Hospital Sírio-Libanês, São Paulo, Brazil
- Division of Radiation Oncology, Department of Radiology and Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Raúl Eduardo Giglio
- Unidad Funcional de Tumore de Cabeza y Cuello, Instituto de Oncología Ángel H. Roffo Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ricard Mesía
- Medical Oncology Department, Catalan Institut of Oncology, 08916 Badalona, Spain and B-ARGO Group, IGTP, Badalona, Spain
| | - Amanda Psyrri
- National Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Assuntina G Sacco
- Division of Hematology-Oncology, Department of Medicine, University of California San Diego Health, Moores Cancer Center, La Jolla, CA, United States
| | - Jatin Shah
- Head and Neck Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Peng Diao
- Department of Radiation Oncology, Sichuan Cancer Hospital, Chengdu, Sichuan, China
| | - Mona Malekzadeh Moghani
- Department of Radiation Oncology, Infertility and Reproductive Health Research Center, Shaid Behesti University of Medical Sciences, Teheran, Iran
| | - Pablo Moreno-Acosta
- Clinical, Molecular and Cellular Radiobiology Research Group, Instituto Nacional de Cancerologia, Bogota, Colombia
| | - Wafa Bouleftour
- Department of Medical Oncology, North Hospital, University Hospital of Saint-Etienne, Saint-Etienne, 42270, France
| | - Eric Deutsch
- Department of Radiotherapy, Université Paris-Saclay, Gustave Roussy, 94805 Villejuif, France and INSERM, Radiothérapie Moléculaire et Innovation Thérapeutique, 94805 Villejuif, France
| |
Collapse
|
6
|
Yu Y, Cai G, Lin R, Wang Z, Chen Y, Tan Y, He Z, Sun Z, Ouyang W, Yao H, Zhang K. Multimodal data fusion AI model uncovers tumor microenvironment immunotyping heterogeneity and enhanced risk stratification of breast cancer. MedComm (Beijing) 2024; 5:e70023. [PMID: 39669975 PMCID: PMC11635117 DOI: 10.1002/mco2.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 08/06/2024] [Accepted: 08/15/2024] [Indexed: 12/14/2024] Open
Abstract
Breast cancer is the leading cancer among women, with a significant number experiencing recurrence and metastasis, thereby reducing survival rates. This study focuses on the role of long noncoding RNAs (lncRNAs) in breast cancer immunotherapy response. We conducted an analysis involving 1027 patients from Sun Yat-sen Memorial Hospital, Sun Yat-sen University, and The Cancer Genome Atlas, utilizing RNA sequencing and pathology whole-slide images. We employed unsupervised clustering to identify distinct lncRNA expression patterns and developed an AI-based pathology model using convolutional neural networks to predict immune-metabolic subtypes. Additionally, we created a multimodal model integrating lncRNA data, immune-cell scores, clinical information, and pathology images for prognostic prediction. Our findings revealed four unique immune-metabolic subtypes, and the AI model demonstrated high predictive accuracy, highlighting the significant impact of lncRNAs on antitumor immunity and metabolic states within the tumor microenvironment. The AI-based pathology model, DeepClinMed-IM, exhibited high accuracy in predicting these subtypes. Additionally, the multimodal model, DeepClinMed-PGM, integrating pathology images, lncRNA data, immune-cell scores, and clinical information, showed superior prognostic performance. In conclusion, these AI models provide a robust foundation for precise prognostication and the identification of potential candidates for immunotherapy, advancing breast cancer research and treatment strategies.
Collapse
Affiliation(s)
- Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical OncologyBreast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Faculty of MedicineMacau University of Science and TechnologyTaipaMacaoChina
| | - Gengyi Cai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical OncologyBreast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Ruichong Lin
- Faculty of Innovation EngineeringMacau University of Science and TechnologyTaipaMacauChina
| | - Zehua Wang
- Faculty of Innovation EngineeringMacau University of Science and TechnologyTaipaMacauChina
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular MedicineKarolinska InstituteStockholmSweden
| | - Yujie Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical OncologyBreast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Zifan He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical OncologyBreast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Zhuo Sun
- Institute for Advanced Study on Eye Health and DiseasesWenzhou Medical UniversityWenzhouChina
| | - Wenhao Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical OncologyBreast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical OncologyBreast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Kang Zhang
- Faculty of MedicineMacau University of Science and TechnologyTaipaMacaoChina
- Faculty of Innovation EngineeringMacau University of Science and TechnologyTaipaMacauChina
- Institute for Advanced Study on Eye Health and DiseasesWenzhou Medical UniversityWenzhouChina
- Guangzhou National LaboratoryGuangzhouChina
- Zhuhai International Eve CenterZhuhai People's Hospital and the First Affiliated Hospital of Faculty of Medicine, Macau University of Science and Technology and University HospitalZhuhaiChina
| |
Collapse
|
7
|
Yu T, Ji Y, Cui X, Liang N, Wu S, Xiang C, Li Y, Tao H, Xie Y, Zuo H, Wang W, Khan N, Ullah K, Xu F, Zhang Y, Lin C. Novel Pathogenic Mutation of P209L in TRPC6 Gene Causes Adult Focal Segmental Glomerulosclerosis. Biochem Genet 2024; 62:4432-4445. [PMID: 38315264 DOI: 10.1007/s10528-023-10651-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 12/27/2023] [Indexed: 02/07/2024]
Abstract
Focal segmental glomerulosclerosis (FSGS) is a leading kidney disease, clinically associated with proteinuria and progressive renal failure. The occurrence of this disease is partly related to gene mutations. We describe a single affected family member who presented with FSGS. We used high-throughput sequencing, sanger sequencing to identify the pathogenic mutations, and a systems genetics analysis in the BXD mice was conducted to explore the genetic regulatory mechanisms of pathogenic genes in the development of FSGS. We identified high urinary protein (++++) and creatinine levels (149 μmol/L) in a 29-year-old male diagnosed with a 5-year history of grade 2 hypertension. Histopathology of the kidney biopsy showed stromal hyperplasia at the glomerular segmental sclerosis and endothelial cell vacuolation degeneration. Whole-exome sequencing followed by Sanger sequencing revealed a heterozygous missense mutation (c.643C > T) in exon 2 of TRPC6, leading to the substitution of arginine with tryptophan at position 215 (p.Arg215Trp). Systems genetics analysis of the 53 BXD mice kidney transcriptomes identified Pygm as the upstream regulator of Trpc6. Those two genes are jointly involved in the regulation of FSGS mainly via Wnt and Hippo signaling pathways. We present a novel variant in the TRPC6 gene that causes FSGS. Moreover, our data suggested TRPC6 works with PYGM, as well as Wnt and Hippo signaling pathways to regulate renal function, which could guide future clinical prevention and targeted treatment for FSGS outcomes.
Collapse
Affiliation(s)
- Tianxi Yu
- School of Clinical Medicine, Weifang Medical University, Weifang, 261042, China
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Yongqiang Ji
- Department of Nephrology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Xin Cui
- School of Clinical Medicine, Weifang Medical University, Weifang, 261042, China
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Ning Liang
- School of Clinical Medicine, Weifang Medical University, Weifang, 261042, China
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Shuang Wu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Chongjun Xiang
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
- The 2nd Medical College of Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Yue Li
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
- The 2nd Medical College of Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Huiying Tao
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
- The 2nd Medical College of Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Yaqi Xie
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
- The 2nd Medical College of Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Hongwei Zuo
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
- The 2nd Medical College of Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Wenting Wang
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Nauman Khan
- Department of Biology, Faculty of Biological and Biomedical Sciences, The University of Haripur, Haripur, KP, Pakistan
| | - Kamran Ullah
- Department of Biology, Faculty of Biological and Biomedical Sciences, The University of Haripur, Haripur, KP, Pakistan
| | - Fuyi Xu
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Yan Zhang
- Department of Nephrology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China.
| | - Chunhua Lin
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China.
| |
Collapse
|
8
|
Abdelaziz EH, Ismail R, Mabrouk MS, Amin E. Multi-omics data integration and analysis pipeline for precision medicine: Systematic review. Comput Biol Chem 2024; 113:108254. [PMID: 39447405 DOI: 10.1016/j.compbiolchem.2024.108254] [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: 06/21/2024] [Revised: 09/05/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
Precision medicine has gained considerable popularity since the "one-size-fits-all" approach did not seem very effective or reflective of the complexity of the human body. Subsequently, since single-omics does not reflect the complexity of the human body's inner workings, it did not result in the expected advancement in the medical field. Therefore, the multi-omics approach has emerged. The multi-omics approach involves integrating data from different omics technologies, such as DNA sequencing, RNA sequencing, mass spectrometry, and others, using computational methods and then analyzing the integrated result for different downstream analysis applications such as survival analysis, cancer classification, or biomarker identification. Most of the recent reviews were constrained to discussing one aspect of the multi-omics analysis pipeline, such as the dimensionality reduction step, the integration methods, or the interpretability aspect; however, very few provide a comprehensive review of every step of the analysis. This study aims to give an overview of the multi-omics analysis pipeline, starting with the most popular multi-omics databases used in recent literature, dimensionality reduction techniques, details the different types of data integration techniques and their downstream analysis applications, describes the most commonly used evaluation metrics, highlights the importance of model interpretability, and lastly discusses the challenges and potential future work for multi-omics data integration in precision medicine.
Collapse
Affiliation(s)
| | - Rasha Ismail
- Faculty of Computer and Information Sciences, Ainshams University, Cairo, Egypt.
| | - Mai S Mabrouk
- Information Technology and Computer Science School, Nile University, Cairo, Egypt.
| | - Eman Amin
- Faculty of Computer and Information Sciences, Ainshams University, Cairo, Egypt.
| |
Collapse
|
9
|
Aziz MA. Multiomics approach towards characterization of tumor cell plasticity and its significance in precision and personalized medicine. Cancer Metastasis Rev 2024; 43:1549-1559. [PMID: 38761231 DOI: 10.1007/s10555-024-10190-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 05/08/2024] [Indexed: 05/20/2024]
Abstract
Cellular plasticity refers to the ability of cells to change their identity or behavior, which can be advantageous in some cases (e.g., tissue regeneration) but detrimental in others (e.g., cancer metastasis). With a better understanding of cellular plasticity, the complexity of cancer cells, their heterogeneity, and their role in metastasis is being unraveled. The plasticity of the cells could also prove as a nemesis to their characterization. In this review, we have attempted to highlight the possibilities and benefits of using multiomics approach in characterizing the plastic nature of cancer cells. There is a need to integrate fragmented evidence at different levels of cellular organization (DNA, RNA, protein, metabolite, epigenetics, etc.) to facilitate the characterization of different forms of plasticity and cell types. We have discussed the role of cellular plasticity in generating intra-tumor heterogeneity. Different omics level evidence is being provided to highlight the variety of molecular determinants discovered using different techniques. Attempts have been made to integrate some of this information to provide a quantitative assessment and scoring of the plastic nature of the cells. However, there is a huge gap in our understanding of mechanisms that lead to the observed heterogeneity. Understanding of these mechanism(s) is necessary for finding targets for early detection and effective therapeutic interventions in metastasis. Targeting cellular plasticity is akin to neutralizing a moving target. Along with the advancements in precision and personalized medicine, these efforts may translate into better clinical outcomes for cancer patients, especially in metastatic stages.
Collapse
Affiliation(s)
- Mohammad Azhar Aziz
- Interdisciplinary Nanotechnology Center, Aligarh Muslim University, Aligarh, Uttar Pradesh, India.
- Cancer Nanomedicine Consortium, Aligarh Muslim University, Aligarh, Uttar Pradesh, India.
| |
Collapse
|
10
|
Xu Y, Cao L, Chen Y, Zhang Z, Liu W, Li H, Ding C, Pu J, Qian K, Xu W. Integrating Machine Learning in Metabolomics: A Path to Enhanced Diagnostics and Data Interpretation. SMALL METHODS 2024; 8:e2400305. [PMID: 38682615 DOI: 10.1002/smtd.202400305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/07/2024] [Indexed: 05/01/2024]
Abstract
Metabolomics, leveraging techniques like NMR and MS, is crucial for understanding biochemical processes in pathophysiological states. This field, however, faces challenges in metabolite sensitivity, data complexity, and omics data integration. Recent machine learning advancements have enhanced data analysis and disease classification in metabolomics. This study explores machine learning integration with metabolomics to improve metabolite identification, data efficiency, and diagnostic methods. Using deep learning and traditional machine learning, it presents advancements in metabolic data analysis, including novel algorithms for accurate peak identification, robust disease classification from metabolic profiles, and improved metabolite annotation. It also highlights multiomics integration, demonstrating machine learning's potential in elucidating biological phenomena and advancing disease diagnostics. This work contributes significantly to metabolomics by merging it with machine learning, offering innovative solutions to analytical challenges and setting new standards for omics data analysis.
Collapse
Affiliation(s)
- Yudian Xu
- Department of Traditional Chinese Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Linlin Cao
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, P. R. China
| | - Yifan Chen
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, P. R. China
| | - Ziyue Zhang
- School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Wanshan Liu
- School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - He Li
- Department of Traditional Chinese Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Chenhuan Ding
- Department of Traditional Chinese Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Jun Pu
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, P. R. China
| | - Kun Qian
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, P. R. China
- School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Wei Xu
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, P. R. China
| |
Collapse
|
11
|
Qi L, Li Z, Liu J, Chen X. Omics-Enhanced Nanomedicine for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409102. [PMID: 39473316 DOI: 10.1002/adma.202409102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/10/2024] [Indexed: 12/13/2024]
Abstract
Cancer nanomedicine has emerged as a promising approach to overcome the limitations of conventional cancer therapies, offering enhanced efficacy and safety in cancer management. However, the inherent heterogeneity of tumors presents increasing challenges for the application of cancer nanomedicine in both diagnosis and treatment. This heterogeneity necessitates the integration of advanced and high-throughput analytical techniques to tailor nanomedicine strategies to individual tumor profiles. Omics technologies, encompassing genomics, epigenomics, transcriptomics, proteomics, metabolomics, and more, provide unparalleled insights into the molecular and cellular mechanisms underlying cancer. By dissecting tumor heterogeneity across multiple levels, these technologies offer robust support for the development of personalized and precise cancer nanomedicine strategies. In this review, the principles, techniques, and applications of key omics technologies are summarized. Especially, the synergistic integration of omics and nanomedicine in cancer therapy is explored, focusing on enhanced diagnostic accuracy, optimized therapeutic strategies and the assessment of nanomedicine-mediated biological responses. Moreover, this review addresses current challenges and outlines future directions in the field of omics-enhanced nanomedicine. By offering valuable insights and guidance, this review aims to advance the integration of omics with nanomedicine, ultimately driving improved diagnostic and therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Lin Qi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, Hunan, 410011, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, Hunan, 410011, China
| | - Jianping Liu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Xiaoyuan Chen
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, Hunan, 410011, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore
| |
Collapse
|
12
|
Li DM, Cao XY, Jiang J. Hypoxia-related bioinformatic signatures associated with prognosis and tumor microenvironment of pancreatic cancer: Current status, concerns, and future perspectives. World J Gastroenterol 2024; 30:4689-4696. [PMID: 39610772 PMCID: PMC11580612 DOI: 10.3748/wjg.v30.i44.4689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/12/2024] Open
Abstract
Pancreatic cancer (PC), a highly lethal tumor with nearly identical incidence and mortality rates, has become the sixth leading cause of cancer-related deaths. Hypoxia is an important malignant factor in PC, as it regulates angiogenesis, metabolic reprogramming, tumor progression, and metastasis. Disrupting the hypoxic microenvironment can enhance the efficacy of antitumor therapy and improve the prognosis of patients with PC. With the advent of bioinformatics, hypoxia-related PC models have emerged in recent years. They provide a reference for estimating the prognosis and immune microenvironment of patients with PC and identify potential biomarkers for targeting hypoxic microenvironment. However, these findings based on bioinformatic analysis may not be completely reliable without further experimental evidence and clinical cohort validation. The application of these models and biomarkers in clinical practice to predict survival time and develop anti hypoxic therapeutic strategies for patients with PC remains in its infancy. In this editorial, we review the current status of hypoxia-related prognostic models in PC, analyze their similarities and differences, discuss several existing challenges, and provide potential solutions and directions for further studies. This editorial will facilitate the optimization, validation, and determination of the molecular mechanisms of related models.
Collapse
Affiliation(s)
- Dong-Ming Li
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Xue-Yuan Cao
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Jing Jiang
- Department of Clinical Epidemiology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
13
|
Wang Y, Wang Z, Yu X, Wang X, Song J, Yu DJ, Ge F. MORE: a multi-omics data-driven hypergraph integration network for biomedical data classification and biomarker identification. Brief Bioinform 2024; 26:bbae658. [PMID: 39692449 DOI: 10.1093/bib/bbae658] [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: 08/19/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
High-throughput sequencing methods have brought about a huge change in omics-based biomedical study. Integrating various omics data is possibly useful for identifying some correlations across data modalities, thus improving our understanding of the underlying biological mechanisms and complexity. Nevertheless, most existing graph-based feature extraction methods overlook the complementary information and correlations across modalities. Moreover, these methods tend to treat the features of each omics modality equally, which contradicts current biological principles. To solve these challenges, we introduce a novel approach for integrating multi-omics data termed Multi-Omics hypeRgraph integration nEtwork (MORE). MORE initially constructs a comprehensive hyperedge group by extensively investigating the informative correlations within and across modalities. Subsequently, the multi-omics hypergraph encoding module is employed to learn the enriched omics-specific information. Afterward, the multi-omics self-attention mechanism is then utilized to adaptatively aggregate valuable correlations across modalities for representation learning and making the final prediction. We assess MORE's performance on datasets characterized by message RNA (mRNA) expression, Deoxyribonucleic Acid (DNA) methylation, and microRNA (miRNA) expression for Alzheimer's disease, invasive breast carcinoma, and glioblastoma. The results from three classification tasks highlight the competitive advantage of MORE in contrast with current state-of-the-art (SOTA) methods. Moreover, the results also show that MORE has the capability to identify a greater variety of disease-related biomarkers compared to existing methods, highlighting its advantages in biomedical data mining and interpretation. Overall, MORE can be investigated as a valuable tool for facilitating multi-omics analysis and novel biomarker discovery. Our code and data can be publicly accessed at https://github.com/Wangyuhanxx/MORE.
Collapse
Affiliation(s)
- Yuhan Wang
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing 210094, China
| | - Zhikang Wang
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Wellington Rd, Clayton, Melbourne, VIC 3800, Australia
| | - Xuan Yu
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong 999077, China
| | - Xiaoyu Wang
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Wellington Rd, Clayton, Melbourne, VIC 3800, Australia
| | - Jiangning Song
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Wellington Rd, Clayton, Melbourne, VIC 3800, Australia
- Monash Data Futures Institute, Monash University, Wellington Rd, Clayton, Melbourne, VIC 3800, Australia
| | - Dong-Jun Yu
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing 210094, China
| | - Fang Ge
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan, Nanjing 210023, China
| |
Collapse
|
14
|
Hamamoto R, Komatsu M, Yamada M, Kobayashi K, Takahashi M, Miyake M, Jinnai S, Koyama T, Kouno N, Machino H, Takahashi S, Asada K, Ueda N, Kaneko S. Current status and future direction of cancer research using artificial intelligence for clinical application. Cancer Sci 2024. [PMID: 39557634 DOI: 10.1111/cas.16395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/23/2024] [Accepted: 11/01/2024] [Indexed: 11/20/2024] Open
Abstract
The expectations for artificial intelligence (AI) technology have increased considerably in recent years, mainly due to the emergence of deep learning. At present, AI technology is being used for various purposes and has brought about change in society. In particular, the rapid development of generative AI technology, exemplified by ChatGPT, has amplified the societal impact of AI. The medical field is no exception, with a wide range of AI technologies being introduced for basic and applied research. Further, AI-equipped software as a medical device (AI-SaMD) is also being approved by regulatory bodies. Combined with the advent of big data, data-driven research utilizing AI is actively pursued. Nevertheless, while AI technology has great potential, it also presents many challenges that require careful consideration. In this review, we introduce the current status of AI-based cancer research, especially from the perspective of clinical application, and discuss the associated challenges and future directions, with the aim of helping to promote cancer research that utilizes effective AI technology.
Collapse
Affiliation(s)
- Ryuji Hamamoto
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Masaaki Komatsu
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Masayoshi Yamada
- Department of Endoscopy, National Cancer Center Hospital, Tokyo, Japan
| | - Kazuma Kobayashi
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Masamichi Takahashi
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan
- Department of Neurosurgery, School of Medicine, Tokai University, Isehara, Kanagawa, Japan
| | - Mototaka Miyake
- Department of Diagnostic Radiology, National Cancer Center Hospital, Tokyo, Japan
| | - Shunichi Jinnai
- Department of Dermatologic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takafumi Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Nobuji Kouno
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidenori Machino
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Satoshi Takahashi
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Ken Asada
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Naonori Ueda
- Disaster Resilience Science Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Syuzo Kaneko
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| |
Collapse
|
15
|
AlDoughaim M, AlSuhebany N, AlZahrani M, AlQahtani T, AlGhamdi S, Badreldin H, Al Alshaykh H. Cancer Biomarkers and Precision Oncology: A Review of Recent Trends and Innovations. Clin Med Insights Oncol 2024; 18:11795549241298541. [PMID: 39559827 PMCID: PMC11571259 DOI: 10.1177/11795549241298541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024] Open
Abstract
The discovery of cancer-specific biomarkers has resulted in major advancements in the field of cancer diagnostics and therapeutics, therefore significantly lowering cancer-related morbidity and mortality. Cancer biomarkers can be generally classified as prognostic biomarkers that predict specific disease outcomes and predictive biomarkers that predict disease response to targeted therapeutic interventions. As research in the area of predictive biomarkers continues to grow, precision medicine becomes far more integrated in cancer treatment. This article presents a general overview on the most recent advancements in the area of cancer biomarkers, immunotherapy, artificial intelligence, and pharmacogenomics of the Middle East.
Collapse
Affiliation(s)
- Maha AlDoughaim
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Nada AlSuhebany
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Mohammed AlZahrani
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Tariq AlQahtani
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Sahar AlGhamdi
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Hisham Badreldin
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Hana Al Alshaykh
- Pharmaceutical Care Devision, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh, Saudi Arabia
| |
Collapse
|
16
|
Li J, Liu Q, Zhang T, Du Q. Bioinformatics Analysis Reveals CDK1 and DLGAP5 as Key Modulators of Tumor Immune Cell Infiltration in Hepatocellular Carcinoma. Cancer Manag Res 2024; 16:1597-1608. [PMID: 39559249 PMCID: PMC11572444 DOI: 10.2147/cmar.s478426] [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: 06/18/2024] [Accepted: 11/01/2024] [Indexed: 11/20/2024] Open
Abstract
Introduction Hepatocellular carcinoma (HCC), a prevalent and aggressive form of cancer, poses significant challenges due to its limited therapeutic options. This study aims to leverage multi-omics data from liver cancer to identify potential therapeutic targets for HCC. Methods We employed an integrative approach by analyzing various omics datasets related to liver cancer. Through comprehensive data mining and analysis, we identified key genes that are significantly associated with HCC. To gain insights into their biological roles and underlying mechanisms, we constructed gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway networks. Specifically, we focused on genes that exhibited high expression levels in HCC and were correlated with poor patient prognosis. Among these, CDK1 and DLGAP5 emerged as promising candidates and were further investigated for their potential involvement in tumor immune cell infiltration and HCC progression. Results Our analysis revealed that CDK1 and DLGAP5 are highly expressed in HCC tissues compared to normal liver tissues, and their elevated expression is associated with unfavorable clinical outcomes. Furthermore, through GO and KEGG pathway analyses, we found that these genes are implicated in critical biological processes and signaling pathways relevant to HCC pathogenesis. Notably, CDK1 and DLGAP5 were shown to be associated with tumor immune cell infiltration, suggesting their potential role in modulating the tumor microenvironment and promoting HCC progression. Discussion These findings provide valuable insights into the development of novel therapeutic approaches for HCC.
Collapse
Affiliation(s)
- Jiajing Li
- The Diagnostics Laboratory, Affiliated Hospital to Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
| | - Qi Liu
- Affiliated Hospital to Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
| | - Ting Zhang
- Affiliated Hospital to Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
| | - Qian Du
- Department of Endoscopy and Digestive System, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, 550002, People’s Republic of China
| |
Collapse
|
17
|
Vashisht V, Vashisht A, Mondal AK, Woodall J, Kolhe R. From Genomic Exploration to Personalized Treatment: Next-Generation Sequencing in Oncology. Curr Issues Mol Biol 2024; 46:12527-12549. [PMID: 39590338 PMCID: PMC11592618 DOI: 10.3390/cimb46110744] [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: 09/04/2024] [Revised: 10/29/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
Next-generation sequencing (NGS) has revolutionized personalized oncology care by providing exceptional insights into the complex genomic landscape. NGS offers comprehensive cancer profiling, which enables clinicians and researchers to better understand the molecular basis of cancer and to tailor treatment strategies accordingly. Targeted therapies based on genomic alterations identified through NGS have shown promise in improving patient outcomes across various cancer types, circumventing resistance mechanisms and enhancing treatment efficacy. Moreover, NGS facilitates the identification of predictive biomarkers and prognostic indicators, aiding in patient stratification and personalized treatment approaches. By uncovering driver mutations and actionable alterations, NGS empowers clinicians to make informed decisions regarding treatment selection and patient management. However, the full potential of NGS in personalized oncology can only be realized through bioinformatics analyses. Bioinformatics plays a crucial role in processing raw sequencing data, identifying clinically relevant variants, and interpreting complex genomic landscapes. This comprehensive review investigates the diverse NGS techniques, including whole-genome sequencing (WGS), whole-exome sequencing (WES), and single-cell RNA sequencing (sc-RNA-Seq), elucidating their roles in understanding the complex genomic/transcriptomic landscape of cancer. Furthermore, the review explores the integration of NGS data with bioinformatics tools to facilitate personalized oncology approaches, from understanding tumor heterogeneity to identifying driver mutations and predicting therapeutic responses. Challenges and future directions in NGS-based cancer research are also discussed, underscoring the transformative impact of these technologies on cancer diagnosis, management, and treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (V.V.); (A.V.); (A.K.M.); (J.W.)
| |
Collapse
|
18
|
Toma MM, Skorski T. Star wars against leukemia: attacking the clones. Leukemia 2024; 38:2293-2302. [PMID: 39223295 PMCID: PMC11519008 DOI: 10.1038/s41375-024-02369-6] [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: 07/02/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Leukemia, although most likely starts as a monoclonal genetic/epigenetic anomaly, is a polyclonal disease at manifestation. This polyclonal nature results from ongoing evolutionary changes in the genome/epigenome of leukemia cells to promote their survival and proliferation advantages. We discuss here how genetic and/or epigenetic aberrations alter intracellular microenvironment in individual leukemia clones and how extracellular microenvironment selects the best fitted clones. This dynamic polyclonal composition of leukemia makes designing an effective therapy a challenging task especially because individual leukemia clones often display substantial differences in response to treatment. Here, we discuss novel therapeutic approach employing single cell multiomics to identify and eradicate all individual clones in a patient.
Collapse
Affiliation(s)
- Monika M Toma
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Tomasz Skorski
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Liu J, Bao C, Zhang J, Han Z, Fang H, Lu H. Artificial intelligence with mass spectrometry-based multimodal molecular profiling methods for advancing therapeutic discovery of infectious diseases. Pharmacol Ther 2024; 263:108712. [PMID: 39241918 DOI: 10.1016/j.pharmthera.2024.108712] [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: 05/31/2024] [Revised: 07/22/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Infectious diseases, driven by a diverse array of pathogens, can swiftly undermine public health systems. Accurate diagnosis and treatment of infectious diseases-centered around the identification of biomarkers and the elucidation of disease mechanisms-are in dire need of more versatile and practical analytical approaches. Mass spectrometry (MS)-based molecular profiling methods can deliver a wealth of information on a range of functional molecules, including nucleic acids, proteins, and metabolites. While MS-driven omics analyses can yield vast datasets, the sheer complexity and multi-dimensionality of MS data can significantly hinder the identification and characterization of functional molecules within specific biological processes and events. Artificial intelligence (AI) emerges as a potent complementary tool that can substantially enhance the processing and interpretation of MS data. AI applications in this context lead to the reduction of spurious signals, the improvement of precision, the creation of standardized analytical frameworks, and the increase of data integration efficiency. This critical review emphasizes the pivotal roles of MS based omics strategies in the discovery of biomarkers and the clarification of infectious diseases. Additionally, the review underscores the transformative ability of AI techniques to enhance the utility of MS-based molecular profiling in the field of infectious diseases by refining the quality and practicality of data produced from omics analyses. In conclusion, we advocate for a forward-looking strategy that integrates AI with MS-based molecular profiling. This integration aims to transform the analytical landscape and the performance of biological molecule characterization, potentially down to the single-cell level. Such advancements are anticipated to propel the development of AI-driven predictive models, thus improving the monitoring of diagnostics and therapeutic discovery for the ongoing challenge related to infectious diseases.
Collapse
Affiliation(s)
- Jingjing Liu
- School of Chinese Medicine, Hong Kong Traditional Chinese Medicine Phenome Research Center, State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong 999077, China
| | - Chaohui Bao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiaxin Zhang
- School of Chinese Medicine, Hong Kong Traditional Chinese Medicine Phenome Research Center, State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong 999077, China
| | - Zeguang Han
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Hai Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Haitao Lu
- School of Chinese Medicine, Hong Kong Traditional Chinese Medicine Phenome Research Center, State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong 999077, China; Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
20
|
Chen X, Yi J, Xie L, Liu T, Liu B, Yan M. Integration of transcriptomics and machine learning for insights into breast cancer: exploring lipid metabolism and immune interactions. Front Immunol 2024; 15:1470167. [PMID: 39524444 PMCID: PMC11543460 DOI: 10.3389/fimmu.2024.1470167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Background Breast cancer (BRCA) represents a substantial global health challenge marked by inadequate early detection rates. The complex interplay between the tumor immune microenvironment and fatty acid metabolism in BRCA requires further investigation to elucidate the specific role of lipid metabolism in this disease. Methods We systematically integrated nine machine learning algorithms into 184 unique combinations to develop a consensus model for lipid metabolism-related prognostic genes (LMPGS). Additionally, transcriptomics analysis provided a comprehensive understanding of this prognostic signature. Using the ESTIMATE method, we evaluated immune infiltration among different risk subgroups and assessed their responsiveness to immunotherapy. Tailored treatments were screened for specific risk subgroups. Finally, we verified the expression of key genes through in vitro experiments. Results We identified 259 differentially expressed genes (DEGs) related to lipid metabolism through analysis of the cancer genome atlas program (TCGA) database. Subsequently, via univariate Cox regression analysis and C-index analysis, we developed an optimal machine learning algorithm to construct a 21-gene LMPGS model. We used optimal cutoff values to divide the lipid metabolism prognostic gene scores into two groups according to high and low scores. Our study revealed distinct biological functions and mutation landscapes between high-scoring and low-scoring patients. The low-scoring group presented a greater immune score, whereas the high-scoring group presented enhanced responses to both immunotherapy and chemotherapy drugs. Single-cell analysis highlighted significant upregulation of CPNE3 in epithelial cells. Moreover, by employing molecular docking, we identified niclosamide as a potential targeted therapeutic drug. Finally, our experiments demonstrated high expression of MTMR9 and CPNE3 in BRCA and their significant correlation with prognosis. Conclusion By employing bioinformatics and diverse machine learning algorithms, we successfully identified genes associated with lipid metabolism in BRCA and uncovered potential therapeutic agents, thereby offering novel insights into the mechanisms and treatment strategies for BRCA.
Collapse
Affiliation(s)
- Xiaohan Chen
- Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Jinfeng Yi
- Department of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Lili Xie
- Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Tong Liu
- Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
- National Health Commission (NHC) Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Baogang Liu
- Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Meisi Yan
- Department of Basic Medical Sciences, Harbin Medical University, Harbin, China
| |
Collapse
|
21
|
Kemkar S, Tao M, Ghosh A, Stamatakos G, Graf N, Poorey K, Balakrishnan U, Trask N, Radhakrishnan R. Towards verifiable cancer digital twins: tissue level modeling protocol for precision medicine. Front Physiol 2024; 15:1473125. [PMID: 39507514 PMCID: PMC11537925 DOI: 10.3389/fphys.2024.1473125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Cancer exhibits substantial heterogeneity, manifesting as distinct morphological and molecular variations across tumors, which frequently undermines the efficacy of conventional oncological treatments. Developments in multiomics and sequencing technologies have paved the way for unraveling this heterogeneity. Nevertheless, the complexity of the data gathered from these methods cannot be fully interpreted through multimodal data analysis alone. Mathematical modeling plays a crucial role in delineating the underlying mechanisms to explain sources of heterogeneity using patient-specific data. Intra-tumoral diversity necessitates the development of precision oncology therapies utilizing multiphysics, multiscale mathematical models for cancer. This review discusses recent advancements in computational methodologies for precision oncology, highlighting the potential of cancer digital twins to enhance patient-specific decision-making in clinical settings. We review computational efforts in building patient-informed cellular and tissue-level models for cancer and propose a computational framework that utilizes agent-based modeling as an effective conduit to integrate cancer systems models that encode signaling at the cellular scale with digital twin models that predict tissue-level response in a tumor microenvironment customized to patient information. Furthermore, we discuss machine learning approaches to building surrogates for these complex mathematical models. These surrogates can potentially be used to conduct sensitivity analysis, verification, validation, and uncertainty quantification, which is especially important for tumor studies due to their dynamic nature.
Collapse
Affiliation(s)
- Sharvari Kemkar
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Mengdi Tao
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Alokendra Ghosh
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Georgios Stamatakos
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, Zografos, Greece
| | - Norbert Graf
- Department of Pediatric Oncology and Hematology, Saarland University, Homburg, Germany
| | - Kunal Poorey
- Department of Systems Biology, Sandia National Laboratories, Livermore, CA, United States
| | - Uma Balakrishnan
- Department of Quant Modeling and SW Eng, Sandia National Laboratories, Livermore, CA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Nathaniel Trask
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA, United States
| | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
22
|
Chhabra R. Molecular and modular intricacies of precision oncology. Front Immunol 2024; 15:1476494. [PMID: 39507541 PMCID: PMC11537923 DOI: 10.3389/fimmu.2024.1476494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Precision medicine is revolutionizing the world in combating different disease modalities, including cancer. The concept of personalized treatments is not new, but modeling it into a reality has faced various limitations. The last decade has seen significant improvements in incorporating several novel tools, scientific innovations and governmental support in precision oncology. However, the socio-economic factors and risk-benefit analyses are important considerations. This mini review includes a summary of some commendable milestones, which are not just a series of successes, but also a cautious outlook to the challenges and practical implications of the advancing techno-medical era.
Collapse
Affiliation(s)
- Ravneet Chhabra
- Business Department, Biocytogen Boston Corporation, Waltham, MA, United States
| |
Collapse
|
23
|
Sun L, Zhang R, Gu Y, Huang L, Jin C. Application of Artificial Intelligence in the diagnosis and treatment of colorectal cancer: a bibliometric analysis, 2004-2023. Front Oncol 2024; 14:1424044. [PMID: 39464716 PMCID: PMC11502294 DOI: 10.3389/fonc.2024.1424044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/23/2024] [Indexed: 10/29/2024] Open
Abstract
Background An increasing number of studies have turned their lens to the application of Artificial Intelligence (AI) in the diagnosis and treatment of colorectal cancer (CRC). Objective To clarify and visualize the basic situation, research hotspots, and development trends of AI in the diagnosis and treatment of CRC, and provide clues for research in the future. Methods On January 31, 2024, the Web of Science Core Collection (WoSCC) database was searched to screen and export the relevant research published during 2004-2023, and Cite Space, VoSviewer, Bibliometrix were used to visualize the number of publications, countries (regions), institutions, journals, authors, citations, keywords, etc. Results A total of 2715 pieces of literature were included. The number of publications grew slowly until the end of 2016, but rapidly after 2017, till to the peak of 798 in 2023. A total of 92 countries, 3997 organizations, and 15,667 authors were involved in this research. Chinese scholars released the highest number of publications, and the U.S. contributed the highest number of total citations. As to authors, MORI, YUICHI had the highest number of publications, and WANG, PU had the highest number of total citations. According to the analysis of citations and keywords, the current research hotspots are mainly related to "Colonoscopy", "Polyp Segmentation", "Digital Pathology", "Radiomics", "prognosis". Conclusion Research on the application of AI in the diagnosis and treatment of CRC has made significant progress and is flourishing across the world. Current research hotspots include AI-assisted early screening and diagnosis, pathology, and staging, and prognosis assessment, and future research is predicted to put weight on multimodal data fusion, personalized treatment, and drug development.
Collapse
Affiliation(s)
- Lamei Sun
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
- Department of Traditional Chinese Medicine, Jiangyin Nanzha Community Health Service Center, Wuxi, China
| | - Rong Zhang
- Department of General Surgery, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Yidan Gu
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Lei Huang
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Chunhui Jin
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
24
|
Salto-Tellez M, Eloy C, Laurinavicius A, Fraggetta F. Integrated diagnostics, complex biomarkers, and a new frontier for tissue pathology. Histopathology 2024; 85:562-565. [PMID: 39210586 DOI: 10.1111/his.15298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024]
Abstract
What will be the next disruptive technology that will change pathology's routine practice again? In this editorial we make a case for the need of more complex biomarkers in oncology diagnostics, to match the inherent complexity of cancer biology. This complexity will be achieved by the validation of technology able to generate more meaningful biological datapoints (epitomized in tissue pathology by technologies such as multiplex immunofluorescence) and, more important, by the systematic analysis of multimodal technology outputs with artificial intelligence tools, which is the essence of integrated diagnostics. While describing these processes, the authors highlight the pivotal role that histopathology will play, once again, in yet another transformation in diagnostics.
Collapse
Affiliation(s)
- Manuel Salto-Tellez
- Integrated Pathology Unit, Division of Molecular Pathology, The Institute of Cancer Research London and The Royal Marsden Hospital, Sutton, UK
- Precision Medicine Centre of Excellence, The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Catarina Eloy
- Department of Pathology, IPATIMUP (Institute of Molecular Pathology and Immunology of University of Porto) & I3S-Instituto de Investigação e Inovação em Saúde & Pathology Department, Medical Faculty of Porto University, Porto, Portugal
| | - Arvydas Laurinavicius
- Institute of Biomedical Sciences of the Faculty of Medicine of Vilnius University & National Center of Pathology affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | | |
Collapse
|
25
|
Coskun A, Ertaylan G, Pusparum M, Van Hoof R, Kaya ZZ, Khosravi A, Zarrabi A. Advancing personalized medicine: Integrating statistical algorithms with omics and nano-omics for enhanced diagnostic accuracy and treatment efficacy. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167339. [PMID: 38986819 DOI: 10.1016/j.bbadis.2024.167339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
Medical laboratory services enable precise measurement of thousands of biomolecules and have become an inseparable part of high-quality healthcare services, exerting a profound influence on global health outcomes. The integration of omics technologies into laboratory medicine has transformed healthcare, enabling personalized treatments and interventions based on individuals' distinct genetic and metabolic profiles. Interpreting laboratory data relies on reliable reference values. Presently, population-derived references are used for individuals, risking misinterpretation due to population heterogeneity, and leading to medical errors. Thus, personalized references are crucial for precise interpretation of individual laboratory results, and the interpretation of omics data should be based on individualized reference values. We reviewed recent advancements in personalized laboratory medicine, focusing on personalized omics, and discussed strategies for implementing personalized statistical approaches in omics technologies to improve global health and concluded that personalized statistical algorithms for interpretation of omics data have great potential to enhance global health. Finally, we demonstrated that the convergence of nanotechnology and omics sciences is transforming personalized laboratory medicine by providing unparalleled diagnostic precision and innovative therapeutic strategies.
Collapse
Affiliation(s)
- Abdurrahman Coskun
- Acibadem University, School of Medicine, Department of Medical Biochemistry, Istanbul, Turkey.
| | - Gökhan Ertaylan
- Unit Health, Environmental Intelligence, Flemish Institute for Technological Research (VITO), Mol 2400, Belgium
| | - Murih Pusparum
- Unit Health, Environmental Intelligence, Flemish Institute for Technological Research (VITO), Mol 2400, Belgium; I-Biostat, Data Science Institute, Hasselt University, Hasselt 3500, Belgium
| | - Rebekka Van Hoof
- Unit Health, Environmental Intelligence, Flemish Institute for Technological Research (VITO), Mol 2400, Belgium
| | - Zelal Zuhal Kaya
- Nisantasi University, School of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey; Graduate School of Biotehnology and Bioengeneering, Yuan Ze University, Taoyuan 320315, Taiwan; Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| |
Collapse
|
26
|
Sanchez I, Rahman R. Radiogenomics as an Integrated Approach to Glioblastoma Precision Medicine. Curr Oncol Rep 2024; 26:1213-1222. [PMID: 39009914 PMCID: PMC11480134 DOI: 10.1007/s11912-024-01580-z] [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] [Accepted: 07/05/2024] [Indexed: 07/17/2024]
Abstract
PURPOSE OF REVIEW Isocitrate dehydrogenase wild-type glioblastoma is the most aggressive primary brain tumour in adults. Its infiltrative nature and heterogeneity confer a dismal prognosis, despite multimodal treatment. Precision medicine is increasingly advocated to improve survival rates in glioblastoma management; however, conventional neuroimaging techniques are insufficient in providing the detail required for accurate diagnosis of this complex condition. RECENT FINDINGS Advanced magnetic resonance imaging allows more comprehensive understanding of the tumour microenvironment. Combining diffusion and perfusion magnetic resonance imaging to create a multiparametric scan enhances diagnostic power and can overcome the unreliability of tumour characterisation by standard imaging. Recent progress in deep learning algorithms establishes their remarkable ability in image-recognition tasks. Integrating these with multiparametric scans could transform the diagnosis and monitoring of patients by ensuring that the entire tumour is captured. As a corollary, radiomics has emerged as a powerful approach to offer insights into diagnosis, prognosis, treatment, and tumour response through extraction of information from radiological scans, and transformation of these tumour characteristics into quantitative data. Radiogenomics, which links imaging features with genomic profiles, has exhibited its ability in characterising glioblastoma, and determining therapeutic response, with the potential to revolutionise management of glioblastoma. The integration of deep learning algorithms into radiogenomic models has established an automated, highly reproducible means to predict glioblastoma molecular signatures, further aiding prognosis and targeted therapy. However, challenges including lack of large cohorts, absence of standardised guidelines and the 'black-box' nature of deep learning algorithms, must first be overcome before this workflow can be applied in clinical practice.
Collapse
Affiliation(s)
- Isabella Sanchez
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Ruman Rahman
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, NG7 2RD, UK.
| |
Collapse
|
27
|
Acharya D, Mukhopadhyay A. A comprehensive review of machine learning techniques for multi-omics data integration: challenges and applications in precision oncology. Brief Funct Genomics 2024; 23:549-560. [PMID: 38600757 DOI: 10.1093/bfgp/elae013] [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: 10/13/2023] [Revised: 03/12/2024] [Accepted: 03/22/2024] [Indexed: 04/12/2024] Open
Abstract
Multi-omics data play a crucial role in precision medicine, mainly to understand the diverse biological interaction between different omics. Machine learning approaches have been extensively employed in this context over the years. This review aims to comprehensively summarize and categorize these advancements, focusing on the integration of multi-omics data, which includes genomics, transcriptomics, proteomics and metabolomics, alongside clinical data. We discuss various machine learning techniques and computational methodologies used for integrating distinct omics datasets and provide valuable insights into their application. The review emphasizes both the challenges and opportunities present in multi-omics data integration, precision medicine and patient stratification, offering practical recommendations for method selection in various scenarios. Recent advances in deep learning and network-based approaches are also explored, highlighting their potential to harmonize diverse biological information layers. Additionally, we present a roadmap for the integration of multi-omics data in precision oncology, outlining the advantages, challenges and implementation difficulties. Hence this review offers a thorough overview of current literature, providing researchers with insights into machine learning techniques for patient stratification, particularly in precision oncology. Contact: anirban@klyuniv.ac.in.
Collapse
Affiliation(s)
- Debabrata Acharya
- Department of Computer Science & Engineering, University of Kalyani, Kalyani-741235, West Bengal, India
| | - Anirban Mukhopadhyay
- Department of Computer Science & Engineering, University of Kalyani, Kalyani-741235, West Bengal, India
| |
Collapse
|
28
|
Jiang B, Bao L, He S, Chen X, Jin Z, Ye Y. Deep learning applications in breast cancer histopathological imaging: diagnosis, treatment, and prognosis. Breast Cancer Res 2024; 26:137. [PMID: 39304962 DOI: 10.1186/s13058-024-01895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
Breast cancer is the most common malignant tumor among women worldwide and remains one of the leading causes of death among women. Its incidence and mortality rates are continuously rising. In recent years, with the rapid advancement of deep learning (DL) technology, DL has demonstrated significant potential in breast cancer diagnosis, prognosis evaluation, and treatment response prediction. This paper reviews relevant research progress and applies DL models to image enhancement, segmentation, and classification based on large-scale datasets from TCGA and multiple centers. We employed foundational models such as ResNet50, Transformer, and Hover-net to investigate the performance of DL models in breast cancer diagnosis, treatment, and prognosis prediction. The results indicate that DL techniques have significantly improved diagnostic accuracy and efficiency, particularly in predicting breast cancer metastasis and clinical prognosis. Furthermore, the study emphasizes the crucial role of robust databases in developing highly generalizable models. Future research will focus on addressing challenges related to data management, model interpretability, and regulatory compliance, ultimately aiming to provide more precise clinical treatment and prognostic evaluation programs for breast cancer patients.
Collapse
Affiliation(s)
- Bitao Jiang
- Department of Hematology and Oncology, Beilun District People's Hospital, Ningbo, 315800, China.
- Department of Hematology and Oncology, Beilun Branch of the First Affiliated Hospital of Zhejiang University, Ningbo, 315800, China.
| | - Lingling Bao
- Department of Hematology and Oncology, Beilun District People's Hospital, Ningbo, 315800, China
- Department of Hematology and Oncology, Beilun Branch of the First Affiliated Hospital of Zhejiang University, Ningbo, 315800, China
| | - Songqin He
- Department of Oncology, The 906th Hospital of the Joint Logistics Force of the Chinese People's Liberation Army, Ningbo, 315100, China
| | - Xiao Chen
- Department of Oncology, The 906th Hospital of the Joint Logistics Force of the Chinese People's Liberation Army, Ningbo, 315100, China
| | - Zhihui Jin
- Department of Hematology and Oncology, Beilun District People's Hospital, Ningbo, 315800, China
- Department of Hematology and Oncology, Beilun Branch of the First Affiliated Hospital of Zhejiang University, Ningbo, 315800, China
| | - Yingquan Ye
- Department of Oncology, The 906th Hospital of the Joint Logistics Force of the Chinese People's Liberation Army, Ningbo, 315100, China.
| |
Collapse
|
29
|
Kamble P, Nagar PR, Bhakhar KA, Garg P, Sobhia ME, Naidu S, Bharatam PV. Cancer pharmacoinformatics: Databases and analytical tools. Funct Integr Genomics 2024; 24:166. [PMID: 39294509 DOI: 10.1007/s10142-024-01445-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
Cancer is a subject of extensive investigation, and the utilization of omics technology has resulted in the generation of substantial volumes of big data in cancer research. Numerous databases are being developed to manage and organize this data effectively. These databases encompass various domains such as genomics, transcriptomics, proteomics, metabolomics, immunology, and drug discovery. The application of computational tools into various core components of pharmaceutical sciences constitutes "Pharmacoinformatics", an emerging paradigm in rational drug discovery. The three major features of pharmacoinformatics include (i) Structure modelling of putative drugs and targets, (ii) Compilation of databases and analysis using statistical approaches, and (iii) Employing artificial intelligence/machine learning algorithms for the discovery of novel therapeutic molecules. The development, updating, and analysis of databases using statistical approaches play a pivotal role in pharmacoinformatics. Multiple software tools are associated with oncoinformatics research. This review catalogs the databases and computational tools related to cancer drug discovery and highlights their potential implications in the pharmacoinformatics of cancer.
Collapse
Affiliation(s)
- Pradnya Kamble
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Prinsa R Nagar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Kaushikkumar A Bhakhar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Prabha Garg
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - M Elizabeth Sobhia
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Srivatsava Naidu
- Center of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Prasad V Bharatam
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India.
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India.
| |
Collapse
|
30
|
Wang R, Wang L, Ren Y, Han X, Liu Z. Editorial: Multidimensionally decoding the impact of tumor heterogeneity on immunotherapy responsiveness in gastrointestinal tumors. Front Immunol 2024; 15:1482206. [PMID: 39290711 PMCID: PMC11405295 DOI: 10.3389/fimmu.2024.1482206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Affiliation(s)
- Ruizhi Wang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Libo Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
31
|
Zhang Y, Cui Y, Hao C, Li Y, He X, Li W, Yu H. Development of the TP53 mutation associated hypopharyngeal squamous cell carcinoma prognostic model through bulk multi-omics sequencing and single-cell sequencing. Braz J Otorhinolaryngol 2024; 91:101499. [PMID: 39341197 PMCID: PMC11466543 DOI: 10.1016/j.bjorl.2024.101499] [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: 04/24/2024] [Revised: 07/30/2024] [Accepted: 08/18/2024] [Indexed: 09/30/2024] Open
Abstract
OBJECTIVE The aim of this study was to construct a prognostic model based on the TP53 mutation to calculate prognostic risk scores of patients with HPSCC. METHODS TP53 mutation and transcriptome data were downloaded from the TCGA databases. Gene expression data from GSE65858, GSE41613, GSE3292, GSE31056, GSE39366, and GSE227156 datasets were downloaded from the GEO database. GSEA, univariate, multivariate Cox analyses, and LASSO analysis were employed to identify key genes and construct the prognostic model. ROC curves were utilized to validate the OS and RFS results obtained from the model. The associations between risk scores with various clinicopathological characteristics and immune scores were analyzed via ggplot2, corrplot package, and GSVA, respectively. Single-cell sequencing data was analyzed via unbiased clustering and SingleR cell annotations. RESULTS Initially, two key genes, POLD2 and POLR2G, were identified and utilized to construct the prognostic model. Samples were divided into different risk groups via the risk scores obtained from the model, with high-risk group samples exhibiting poorer prognosis. Furthermore, the risk score exhibited a positive correlation with lymphatic metastasis in patients and the immune scores of CD4+ T, CD8+ T, dendritic cell, macrophage, and neutrophil. The immune responses also exhibited notable disparities between the high- and low-risk groups. The results of single-cell sequencing analysis demonstrated that epithelial cells and macrophages were relatively abundant in HPSCC samples. POLD2 and POLR2G exhibited higher expressions in epithelial cells, with most of the identified pathways also enriched in epithelial cells. CONCLUSION The prognostic model exhibited a significant capacity for predicting the prognosis of HSPCC samples based on the TP53 mutation conditions and may also predict the cancer characteristics and immune infiltration scores of samples via different risk scores obtained from the model. LEVEL OF EVIDENCE Level 5.
Collapse
Affiliation(s)
- Ying Zhang
- The Second Affiliated Hospital of Harbin Medical University, Department of Radiation Oncology, Harbin, China
| | - Yue Cui
- The Second Affiliated Hospital of Harbin Medical University, Department of Radiation Oncology, Harbin, China
| | - Congfan Hao
- The Second Affiliated Hospital of Harbin Medical University, Department of Radiation Oncology, Harbin, China
| | - Yingjie Li
- The Second Affiliated Hospital of Harbin Medical University, Department of Radiation Oncology, Harbin, China
| | - Xinyang He
- The Second Affiliated Hospital of Harbin Medical University, Department of Radiation Oncology, Harbin, China
| | - Wenhui Li
- The Second Affiliated Hospital of Harbin Medical University, Department of Radiation Oncology, Harbin, China
| | - Hongyang Yu
- The Second Affiliated Hospital of Harbin Medical University, Department of Radiation Oncology, Harbin, China.
| |
Collapse
|
32
|
Jones CH, Madhavan S, Natarajan K, Corbo M, True JM, Dolsten M. Rewriting the textbook for pharma: how to adapt and thrive in a digital, personalized and collaborative world. Drug Discov Today 2024; 29:104112. [PMID: 39053620 DOI: 10.1016/j.drudis.2024.104112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/01/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
The pharmaceutical industry is undergoing a sweeping transformation, driven by technological innovations, demographic shifts, regulatory changes and consumer expectations. For adaptive players in pharma to excel in this rapidly changing landscape, which will be markedly different from today by 2030 and beyond, they will require a different set of skills, capabilities and mindsets, as well as a willingness to collaborate and co-create value with multiple stakeholders. The industry needs to rewrite the textbook for pharma by embracing and implementing four key dimensions of change: digitalization, personalization, collaboration and innovation. In this article, we will examine how these dimensions of change are reshaping the industry, and provide practical and strategic guidance based on best practices and examples. Specifically, adaptive pharma companies should embrace the use of advanced digital technologies, such as artificial intelligence and machine learning, to streamline processes and solve challenges rapidly. Personalization, both in medicine and patient engagement, will also be key to success in the 'digital revolution', and a collaborative approach involving partnerships with tech start-ups, health-care providers and regulatory bodies will also be essential to create an integrated and responsive health-care ecosystem. Using these ideas for a rewritten textbook for pharma, adaptive players in pharma will evolve to be personalized and digitized health-focused organizations that provide comprehensive solutions which go beyond drugs and devices.
Collapse
Affiliation(s)
| | | | | | - Michael Corbo
- Pfizer, 66 Hudson Boulevard, New York, NY 10018, USA
| | - Jane M True
- Pfizer, 66 Hudson Boulevard, New York, NY 10018, USA
| | | |
Collapse
|
33
|
Zheng Y, Ma Y, Xiong Q, Zhu K, Weng N, Zhu Q. The role of artificial intelligence in the development of anticancer therapeutics from natural polyphenols: Current advances and future prospects. Pharmacol Res 2024; 208:107381. [PMID: 39218422 DOI: 10.1016/j.phrs.2024.107381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/06/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Natural polyphenols, abundant in the human diet, are derived from a wide variety of sources. Numerous preclinical studies have demonstrated their significant anticancer properties against various malignancies, making them valuable resources for drug development. However, traditional experimental methods for developing anticancer therapies from natural polyphenols are time-consuming and labor-intensive. Recently, artificial intelligence has shown promising advancements in drug discovery. Integrating AI technologies into the development process for natural polyphenols can substantially reduce development time and enhance efficiency. In this study, we review the crucial roles of natural polyphenols in anticancer treatment and explore the potential of AI technologies to aid in drug development. Specifically, we discuss the application of AI in key stages such as drug structure prediction, virtual drug screening, prediction of biological activity, and drug-target protein interaction, highlighting the potential to revolutionize the development of natural polyphenol-based anticancer therapies.
Collapse
Affiliation(s)
- Ying Zheng
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Yifei Ma
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Qunli Xiong
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Kai Zhu
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian 350011, PR China
| | - Ningna Weng
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian 350011, PR China
| | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China.
| |
Collapse
|
34
|
Ozcelik F, Dundar MS, Yildirim AB, Henehan G, Vicente O, Sánchez-Alcázar JA, Gokce N, Yildirim DT, Bingol NN, Karanfilska DP, Bertelli M, Pojskic L, Ercan M, Kellermayer M, Sahin IO, Greiner-Tollersrud OK, Tan B, Martin D, Marks R, Prakash S, Yakubi M, Beccari T, Lal R, Temel SG, Fournier I, Ergoren MC, Mechler A, Salzet M, Maffia M, Danalev D, Sun Q, Nei L, Matulis D, Tapaloaga D, Janecke A, Bown J, Cruz KS, Radecka I, Ozturk C, Nalbantoglu OU, Sag SO, Ko K, Arngrimsson R, Belo I, Akalin H, Dundar M. The impact and future of artificial intelligence in medical genetics and molecular medicine: an ongoing revolution. Funct Integr Genomics 2024; 24:138. [PMID: 39147901 DOI: 10.1007/s10142-024-01417-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
Artificial intelligence (AI) platforms have emerged as pivotal tools in genetics and molecular medicine, as in many other fields. The growth in patient data, identification of new diseases and phenotypes, discovery of new intracellular pathways, availability of greater sets of omics data, and the need to continuously analyse them have led to the development of new AI platforms. AI continues to weave its way into the fabric of genetics with the potential to unlock new discoveries and enhance patient care. This technology is setting the stage for breakthroughs across various domains, including dysmorphology, rare hereditary diseases, cancers, clinical microbiomics, the investigation of zoonotic diseases, omics studies in all medical disciplines. AI's role in facilitating a deeper understanding of these areas heralds a new era of personalised medicine, where treatments and diagnoses are tailored to the individual's molecular features, offering a more precise approach to combating genetic or acquired disorders. The significance of these AI platforms is growing as they assist healthcare professionals in the diagnostic and treatment processes, marking a pivotal shift towards more informed, efficient, and effective medical practice. In this review, we will explore the range of AI tools available and show how they have become vital in various sectors of genomic research supporting clinical decisions.
Collapse
Affiliation(s)
- Firat Ozcelik
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Mehmet Sait Dundar
- Department of Electrical and Computer Engineering, Graduate School of Engineering and Sciences, Abdullah Gul University, Kayseri, Turkey
| | - A Baki Yildirim
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Gary Henehan
- School of Food Science and Environmental Health, Technological University of Dublin, Dublin, Ireland
| | - Oscar Vicente
- Institute for the Conservation and Improvement of Valencian Agrodiversity (COMAV), Universitat Politècnica de València, Valencia, Spain
| | - José A Sánchez-Alcázar
- Centro de Investigación Biomédica en Red: Enfermedades Raras, Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Instituto de Salud Carlos III, Sevilla, Spain
| | - Nuriye Gokce
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Duygu T Yildirim
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Nurdeniz Nalbant Bingol
- Department of Translational Medicine, Institute of Health Sciences, Bursa Uludag University, Bursa, Turkey
| | - Dijana Plaseska Karanfilska
- Research Centre for Genetic Engineering and Biotechnology, Macedonian Academy of Sciences and Arts, Skopje, Macedonia
| | | | - Lejla Pojskic
- Institute for Genetic Engineering and Biotechnology, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Mehmet Ercan
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Miklos Kellermayer
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Izem Olcay Sahin
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | | | - Busra Tan
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Donald Martin
- University Grenoble Alpes, CNRS, TIMC-IMAG/SyNaBi (UMR 5525), Grenoble, France
| | - Robert Marks
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Satya Prakash
- Department of Biomedical Engineering, University of McGill, Montreal, QC, Canada
| | - Mustafa Yakubi
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Tommaso Beccari
- Department of Pharmeceutical Sciences, University of Perugia, Perugia, Italy
| | - Ratnesh Lal
- Neuroscience Research Institute, University of California, Santa Barbara, USA
| | - Sehime G Temel
- Department of Translational Medicine, Institute of Health Sciences, Bursa Uludag University, Bursa, Turkey
- Department of Medical Genetics, Bursa Uludag University Faculty of Medicine, Bursa, Turkey
- Department of Histology and Embryology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Isabelle Fournier
- Réponse Inflammatoire et Spectrométrie de Masse-PRISM, University of Lille, Lille, France
| | - M Cerkez Ergoren
- Department of Medical Genetics, Near East University Faculty of Medicine, Nicosia, Cyprus
| | - Adam Mechler
- Department of Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Michel Salzet
- Réponse Inflammatoire et Spectrométrie de Masse-PRISM, University of Lille, Lille, France
| | - Michele Maffia
- Department of Experimental Medicine, University of Salento, Via Lecce-Monteroni, Lecce, 73100, Italy
| | - Dancho Danalev
- University of Chemical Technology and Metallurgy, Sofia, Bulgaria
| | - Qun Sun
- Department of Food Science and Technology, Sichuan University, Chengdu, China
| | - Lembit Nei
- School of Engineering Tallinn University of Technology, Tartu College, Tartu, Estonia
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Dana Tapaloaga
- Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine of Bucharest, Bucharest, Romania
| | - Andres Janecke
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
- Division of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - James Bown
- School of Science, Engineering and Technology, Abertay University, Dundee, UK
| | | | - Iza Radecka
- School of Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, UK
| | - Celal Ozturk
- Department of Software Engineering, Erciyes University, Kayseri, Turkey
| | - Ozkan Ufuk Nalbantoglu
- Department of Computer Engineering, Engineering Faculty, Erciyes University, Kayseri, Turkey
| | - Sebnem Ozemri Sag
- Department of Medical Genetics, Bursa Uludag University Faculty of Medicine, Bursa, Turkey
| | - Kisung Ko
- Department of Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Reynir Arngrimsson
- Iceland Landspitali University Hospital, University of Iceland, Reykjavik, Iceland
| | - Isabel Belo
- Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Hilal Akalin
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey.
| | - Munis Dundar
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey.
| |
Collapse
|
35
|
Isavand P, Aghamiri SS, Amin R. Applications of Multimodal Artificial Intelligence in Non-Hodgkin Lymphoma B Cells. Biomedicines 2024; 12:1753. [PMID: 39200217 PMCID: PMC11351272 DOI: 10.3390/biomedicines12081753] [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: 06/25/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
Given advancements in large-scale data and AI, integrating multimodal artificial intelligence into cancer research can enhance our understanding of tumor behavior by simultaneously processing diverse biomedical data types. In this review, we explore the potential of multimodal AI in comprehending B-cell non-Hodgkin lymphomas (B-NHLs). B-cell non-Hodgkin lymphomas (B-NHLs) represent a particular challenge in oncology due to tumor heterogeneity and the intricate ecosystem in which tumors develop. These complexities complicate diagnosis, prognosis, and therapy response, emphasizing the need to use sophisticated approaches to enhance personalized treatment strategies for better patient outcomes. Therefore, multimodal AI can be leveraged to synthesize critical information from available biomedical data such as clinical record, imaging, pathology and omics data, to picture the whole tumor. In this review, we first define various types of modalities, multimodal AI frameworks, and several applications in precision medicine. Then, we provide several examples of its usage in B-NHLs, for analyzing the complexity of the ecosystem, identifying immune biomarkers, optimizing therapy strategy, and its clinical applications. Lastly, we address the limitations and future directions of multimodal AI, highlighting the need to overcome these challenges for better clinical practice and application in healthcare.
Collapse
Affiliation(s)
- Pouria Isavand
- Department of Radiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan 4513956184, Iran
| | | | - Rada Amin
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68503, USA
| |
Collapse
|
36
|
Wu S, Sun X, Hua R, Hu C, Qin L. DDX21 functions as a potential novel oncopromoter in pancreatic ductal adenocarcinoma: a comprehensive analysis of the DExD box family. Discov Oncol 2024; 15:333. [PMID: 39095628 PMCID: PMC11297014 DOI: 10.1007/s12672-024-01204-9] [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: 06/10/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal tumor with an ill-defined pathogenesis. DExD box (DDX) family genes are widely distributed and involved in various RNA metabolism and cellular biogenesis; their dysregulation is associated with aberrant cellular processes and malignancies. However, the prognostic significance and expression patterns of the DDX family in PDAC are not fully understood. The present study aimed to explore the clinical value of DDX genes in PDAC. METHODS Differentially expressed DDX genes were identified. DDX genes related to prognostic signatures were further investigated using LASSO Cox regression analysis. DDX21 protein expression was analyzed using the UALCAN and human protein atlas (HPA) online tools and confirmed in 40 paired PDAC and normal tissues through Tissue Microarrays (TMA). The independent prognostic significance of DDX21 in PDAC was determined through the construction of nomogram models and calibration curves. The functional roles of DDX21 were investigated using gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA). Cell proliferation, invasion, and migration were assessed using Cell Counting Kit-8, colony formation, Transwell, and wound healing assays. RESULTS Upregulation of genes related to prognostic signatures (DDX10, DDX21, DDX60, and DDX60L) was significantly associated with poor prognosis of patients with PDAC based on survival and recurrence time. Considering the expression profile and prognostic values of the signature-related genes, DDX21 was finally selected for further exploration. DDX21 was overexpressed significantly at both the mRNA and protein levels in PDAC compared to normal pancreatic tissues. DDX21 expression, pathological stage, and residual tumor were significant independent prognostic indicators in PDAC. Moreover, functional enrichment analysis revealed that Genes co-expressed with DDX21 are predominantly involved in RNA metabolism, helicase activity, ribosome biogenesis, cell cycle, and various cancer-related pathways, such as PI3K/Akt signaling pathway and TGF-β signaling pathway. Furthermore, in vitro experiments confirmed that the knockdown of DDX21 significantly reduced MIA PaCa-2 cell viability, proliferation, migration, and invasion. CONCLUSIONS Four signature-related genes could relatively precisely predict the prognosis of patients with PDAC. Specifically, DDX21 upregulation may signal an unfavorable prognosis by negatively affecting the biological properties of PDAC cells. DDX21 may be considered as a candidate therapeutic target in PDAC.
Collapse
Affiliation(s)
- Shaohan Wu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 899, Pinghai Road, Suzhou, 215006, Jiangsu, China
- Department of General Surgery, the Second Affiliated Hospital of Jiaxing University, No. 1518, Huancheng North Road, Jiaxing, 314000, Zhejiang, China
| | - Xiaofang Sun
- Department of General Surgery, the Second Affiliated Hospital of Jiaxing University, No. 1518, Huancheng North Road, Jiaxing, 314000, Zhejiang, China
| | - Ruheng Hua
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 899, Pinghai Road, Suzhou, 215006, Jiangsu, China
| | - Chundong Hu
- Department of General Surgery, the Second Affiliated Hospital of Jiaxing University, No. 1518, Huancheng North Road, Jiaxing, 314000, Zhejiang, China
| | - Lei Qin
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 899, Pinghai Road, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
37
|
Loaiza RA, Farías MA, Andrade CA, Ramírez MA, Rodriguez-Guilarte L, Muñóz JT, González PA, Bueno SM, Kalergis AM. Immunomodulatory markers and therapies for the management of infant respiratory syncytial virus infection. Expert Rev Anti Infect Ther 2024; 22:631-645. [PMID: 39269198 DOI: 10.1080/14787210.2024.2403147] [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: 06/04/2024] [Revised: 08/16/2024] [Accepted: 09/08/2024] [Indexed: 09/15/2024]
Abstract
INTRODUCTION The human respiratory syncytial virus (hRSV) is one of childhood diseases' most common respiratory pathogens and is associated with lower respiratory tract infections. The peak in disease that this virus can elicit during outbreaks is often a significant burden for healthcare systems worldwide. Despite theapproval of treatments against hRSV, this pathogen remains one the most common causative agent of infant mortality around the world. AREAS COVERED This review focuses on the key prognostic and immunomodulatory biomarkers associated with hRSV infection, as well as prophylactic monoclonal antibodies and vaccines. The goal is to catalyze a paradigm shift within the scientific community toward the discovery of novel targets to predict the clinical outcome of infected patients, as well as the development of novel antiviral agents targeting hRSV. The most pertinent research on this topic was systematically searched and analyzed using PubMed ISI Thomson Scientific databases. EXPERT OPINION Despite advances in approved therapies against hRSV, it is crucial to continue researching to develop new therapies and to find specific biomarkers to predict the severity of infection. Along these lines, the use of multi-omics data, artificial intelligence and natural-derived compounds with antiviral activity could be evaluated to fight hRSV and develop methods for rapid diagnosis of severity.
Collapse
Affiliation(s)
- Ricardo A Loaiza
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A Andrade
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mario A Ramírez
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Linmar Rodriguez-Guilarte
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José T Muñóz
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
38
|
Jiang H, Liu M, Deng Y, Zhang C, Dai L, Zhu B, Ou Y, Zhu Y, Hu C, Yang L, Li J, Bai Y, Yang D. Identification of prostate cancer bone metastasis related genes and potential therapy targets by bioinformatics and in vitro experiments. J Cell Mol Med 2024; 28:e18511. [PMID: 39098992 PMCID: PMC11298316 DOI: 10.1111/jcmm.18511] [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: 04/23/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 08/06/2024] Open
Abstract
The aetiology of bone metastasis in prostate cancer (PCa) remains unclear. This study aims to identify hub genes involved in this process. We utilized machine learning, GO, KEGG, GSEA, Single-cell analysis, ROC methods to identify hub genes for bone metastasis in PCa using the TCGA and GEO databases. Potential drugs targeting these genes were identified. We validated these results using 16 specimens from patients with PCa and analysed the relationship between the hub genes and clinical features. The impact of APOC1 on PCa was assessed through in vitro experiments. Seven hub genes with AUC values of 0.727-0.926 were identified. APOC1, CFH, NUSAP1 and LGALS1 were highly expressed in bone metastasis tissues, while NR4A2, ADRB2 and ZNF331 exhibited an opposite trend. Immunohistochemistry further confirmed these results. The oxidative phosphorylation pathway was significantly enriched by the identified genes. Aflatoxin B1, benzo(a)pyrene, cyclosporine were identified as potential drugs. APOC1 expression was correlated with clinical features of PCa metastasis. Silencing APOC1 significantly inhibited PCa cell proliferation, clonality, and migration in vitro. This study identified 7 hub genes that potentially facilitate bone metastasis in PCa through mitochondrial metabolic reprogramming. APOC1 emerged as a promising therapeutic target and prognostic marker for PCa with bone metastasis.
Collapse
Affiliation(s)
- Haiyang Jiang
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
- Department of Urology IIThe second Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Mingcheng Liu
- Department of Human Cell Biology and Genetics, School of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Yingfei Deng
- Pathology‐DepartmentThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Chongjian Zhang
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Longguo Dai
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Bingyu Zhu
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Yitian Ou
- Department of Urology IIThe second Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Yong Zhu
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Chen Hu
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Libo Yang
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Jun Li
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Yu Bai
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Delin Yang
- Department of Urology IIThe second Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| |
Collapse
|
39
|
Zhao Y, Li X, Zhou C, Peng H, Zheng Z, Chen J, Ding W. A review of cancer data fusion methods based on deep learning. INFORMATION FUSION 2024; 108:102361. [DOI: 10.1016/j.inffus.2024.102361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
40
|
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
|
41
|
Li L, Sun M, Wang J, Wan S. Multi-omics based artificial intelligence for cancer research. Adv Cancer Res 2024; 163:303-356. [PMID: 39271266 DOI: 10.1016/bs.acr.2024.06.005] [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] [Indexed: 09/15/2024]
Abstract
With significant advancements of next generation sequencing technologies, large amounts of multi-omics data, including genomics, epigenomics, transcriptomics, proteomics, and metabolomics, have been accumulated, offering an unprecedented opportunity to explore the heterogeneity and complexity of cancer across various molecular levels and scales. One of the promising aspects of multi-omics lies in its capacity to offer a holistic view of the biological networks and pathways underpinning cancer, facilitating a deeper understanding of its development, progression, and response to treatment. However, the exponential growth of data generated by multi-omics studies present significant analytical challenges. Processing, analyzing, integrating, and interpreting these multi-omics datasets to extract meaningful insights is an ambitious task that stands at the forefront of current cancer research. The application of artificial intelligence (AI) has emerged as a powerful solution to these challenges, demonstrating exceptional capabilities in deciphering complex patterns and extracting valuable information from large-scale, intricate omics datasets. This review delves into the synergy of AI and multi-omics, highlighting its revolutionary impact on oncology. We dissect how this confluence is reshaping the landscape of cancer research and clinical practice, particularly in the realms of early detection, diagnosis, prognosis, treatment and pathology. Additionally, we elaborate the latest AI methods for multi-omics integration to provide a comprehensive insight of the complex biological mechanisms and inherent heterogeneity of cancer. Finally, we discuss the current challenges of data harmonization, algorithm interpretability, and ethical considerations. Addressing these challenges necessitates a multidisciplinary collaboration, paving the promising way for more precise, personalized, and effective treatments for cancer patients.
Collapse
Affiliation(s)
- Lusheng Li
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mengtao Sun
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jieqiong Wang
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shibiao Wan
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
42
|
Mohr AE, Ortega-Santos CP, Whisner CM, Klein-Seetharaman J, Jasbi P. Navigating Challenges and Opportunities in Multi-Omics Integration for Personalized Healthcare. Biomedicines 2024; 12:1496. [PMID: 39062068 PMCID: PMC11274472 DOI: 10.3390/biomedicines12071496] [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: 04/15/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
The field of multi-omics has witnessed unprecedented growth, converging multiple scientific disciplines and technological advances. This surge is evidenced by a more than doubling in multi-omics scientific publications within just two years (2022-2023) since its first referenced mention in 2002, as indexed by the National Library of Medicine. This emerging field has demonstrated its capability to provide comprehensive insights into complex biological systems, representing a transformative force in health diagnostics and therapeutic strategies. However, several challenges are evident when merging varied omics data sets and methodologies, interpreting vast data dimensions, streamlining longitudinal sampling and analysis, and addressing the ethical implications of managing sensitive health information. This review evaluates these challenges while spotlighting pivotal milestones: the development of targeted sampling methods, the use of artificial intelligence in formulating health indices, the integration of sophisticated n-of-1 statistical models such as digital twins, and the incorporation of blockchain technology for heightened data security. For multi-omics to truly revolutionize healthcare, it demands rigorous validation, tangible real-world applications, and smooth integration into existing healthcare infrastructures. It is imperative to address ethical dilemmas, paving the way for the realization of a future steered by omics-informed personalized medicine.
Collapse
Affiliation(s)
- Alex E. Mohr
- Systems Precision Engineering and Advanced Research (SPEAR), Theriome Inc., Phoenix, AZ 85004, USA; (A.E.M.); (C.P.O.-S.); (C.M.W.); (J.K.-S.)
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
- Biodesign Institute Center for Health Through Microbiomes, Arizona State University, Tempe, AZ 85281, USA
| | - Carmen P. Ortega-Santos
- Systems Precision Engineering and Advanced Research (SPEAR), Theriome Inc., Phoenix, AZ 85004, USA; (A.E.M.); (C.P.O.-S.); (C.M.W.); (J.K.-S.)
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, George Washington University, Washington, DC 20052, USA
| | - Corrie M. Whisner
- Systems Precision Engineering and Advanced Research (SPEAR), Theriome Inc., Phoenix, AZ 85004, USA; (A.E.M.); (C.P.O.-S.); (C.M.W.); (J.K.-S.)
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
- Biodesign Institute Center for Health Through Microbiomes, Arizona State University, Tempe, AZ 85281, USA
| | - Judith Klein-Seetharaman
- Systems Precision Engineering and Advanced Research (SPEAR), Theriome Inc., Phoenix, AZ 85004, USA; (A.E.M.); (C.P.O.-S.); (C.M.W.); (J.K.-S.)
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85281, USA
| | - Paniz Jasbi
- Systems Precision Engineering and Advanced Research (SPEAR), Theriome Inc., Phoenix, AZ 85004, USA; (A.E.M.); (C.P.O.-S.); (C.M.W.); (J.K.-S.)
| |
Collapse
|
43
|
Khera R, Oikonomou EK, Nadkarni GN, Morley JR, Wiens J, Butte AJ, Topol EJ. Transforming Cardiovascular Care With Artificial Intelligence: From Discovery to Practice: JACC State-of-the-Art Review. J Am Coll Cardiol 2024; 84:97-114. [PMID: 38925729 DOI: 10.1016/j.jacc.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 06/28/2024]
Abstract
Artificial intelligence (AI) has the potential to transform every facet of cardiovascular practice and research. The exponential rise in technology powered by AI is defining new frontiers in cardiovascular care, with innovations that span novel diagnostic modalities, new digital native biomarkers of disease, and high-performing tools evaluating care quality and prognosticating clinical outcomes. These digital innovations promise expanded access to cardiovascular screening and monitoring, especially among those without access to high-quality, specialized care historically. Moreover, AI is propelling biological and clinical discoveries that will make future cardiovascular care more personalized, precise, and effective. The review brings together these diverse AI innovations, highlighting developments in multimodal cardiovascular AI across clinical practice and biomedical discovery, and envisioning this new future backed by contemporary science and emerging discoveries. Finally, we define the critical path and the safeguards essential to realizing this AI-enabled future that helps achieve optimal cardiovascular health and outcomes for all.
Collapse
Affiliation(s)
- Rohan Khera
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA; Center for Outcomes Research and Evaluation (CORE), New Haven, Connecticut, USA; Section of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, Connecticut, USA; Section of Health Informatics, Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA.
| | - Evangelos K Oikonomou
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Girish N Nadkarni
- The Samuel Bronfman Department of Medicine, Division of Data Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, New York, USA; The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jessica R Morley
- Digital Ethics Center, Yale University, New Haven, Connecticut, USA
| | - Jenna Wiens
- Electrical Engineering and Computer Science, Computer Science and Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Atul J Butte
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, California, USA; Center for Data-Driven Insights and Innovation, University of California Health, Oakland, California, USA
| | - Eric J Topol
- Molecular Medicine, Scripps Research Translational Institute, Scripps Research, La Jolla, California, USA
| |
Collapse
|
44
|
Nussinov R, Yavuz BR, Demirel HC, Arici MK, Jang H, Tuncbag N. Review: Cancer and neurodevelopmental disorders: multi-scale reasoning and computational guide. Front Cell Dev Biol 2024; 12:1376639. [PMID: 39015651 PMCID: PMC11249571 DOI: 10.3389/fcell.2024.1376639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
The connection and causality between cancer and neurodevelopmental disorders have been puzzling. How can the same cellular pathways, proteins, and mutations lead to pathologies with vastly different clinical presentations? And why do individuals with neurodevelopmental disorders, such as autism and schizophrenia, face higher chances of cancer emerging throughout their lifetime? Our broad review emphasizes the multi-scale aspect of this type of reasoning. As these examples demonstrate, rather than focusing on a specific organ system or disease, we aim at the new understanding that can be gained. Within this framework, our review calls attention to computational strategies which can be powerful in discovering connections, causalities, predicting clinical outcomes, and are vital for drug discovery. Thus, rather than centering on the clinical features, we draw on the rapidly increasing data on the molecular level, including mutations, isoforms, three-dimensional structures, and expression levels of the respective disease-associated genes. Their integrated analysis, together with chromatin states, can delineate how, despite being connected, neurodevelopmental disorders and cancer differ, and how the same mutations can lead to different clinical symptoms. Here, we seek to uncover the emerging connection between cancer, including pediatric tumors, and neurodevelopmental disorders, and the tantalizing questions that this connection raises.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, United States
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, United States
| | | | - M. Kaan Arici
- Graduate School of Informatics, Middle East Technical University, Ankara, Türkiye
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, United States
| | - Nurcan Tuncbag
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Türkiye
- School of Medicine, Koc University, Istanbul, Türkiye
- Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| |
Collapse
|
45
|
Wang H, Li X, Shi P, You X, Zhao G. Establishment and evaluation of on-chip intestinal barrier biosystems based on microfluidic techniques. Mater Today Bio 2024; 26:101079. [PMID: 38774450 PMCID: PMC11107260 DOI: 10.1016/j.mtbio.2024.101079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/17/2024] [Accepted: 05/01/2024] [Indexed: 05/24/2024] Open
Abstract
As a booming engineering technology, the microfluidic chip has been widely applied for replicating the complexity of human intestinal micro-physiological ecosystems in vitro. Biosensors, 3D imaging, and multi-omics have been applied to engineer more sophisticated intestinal barrier-on-chip platforms, allowing the improved monitoring of physiological processes and enhancing chip performance. In this review, we report cutting-edge advances in the microfluidic techniques applied for the establishment and evaluation of intestinal barrier platforms. We discuss different design principles and microfabrication strategies for the establishment of microfluidic gut barrier models in vitro. Further, we comprehensively cover the complex cell types (e.g., epithelium, intestinal organoids, endothelium, microbes, and immune cells) and controllable extracellular microenvironment parameters (e.g., oxygen gradient, peristalsis, bioflow, and gut-organ axis) used to recapitulate the main structural and functional complexity of gut barriers. We also present the current multidisciplinary technologies and indicators used for evaluating the morphological structure and barrier integrity of established gut barrier models in vitro. Finally, we highlight the challenges and future perspectives for accelerating the broader applications of these platforms in disease simulation, drug development, and personalized medicine. Hence, this review provides a comprehensive guide for the development and evaluation of microfluidic-based gut barrier platforms.
Collapse
Affiliation(s)
- Hui Wang
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, China
| | - Xiangyang Li
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, 471023, China
- Haihe Laboratory of Synthetic Biology, Tianjin, 300308, China
| | - Pengcheng Shi
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, 471023, China
| | - Xiaoyan You
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, China
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, 471023, China
| | - Guoping Zhao
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
- CAS-Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
46
|
Zhou Z, Lin T, Chen S, Zhang G, Xu Y, Zou H, Zhou A, Zhang Y, Weng S, Han X, Liu Z. Omics-based molecular classifications empowering in precision oncology. Cell Oncol (Dordr) 2024; 47:759-777. [PMID: 38294647 DOI: 10.1007/s13402-023-00912-8] [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] [Accepted: 12/23/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND In the past decades, cancer enigmatical heterogeneity at distinct expression levels could interpret disparities in therapeutic response and prognosis. It built hindrances to precision medicine, a tactic to tailor customized treatment informed by the tumors' molecular profile. Single-omics analysis dissected the biological features associated with carcinogenesis to some extent but still failed to revolutionize cancer treatment as expected. Integrated omics analysis incorporated tumor biological networks from diverse layers and deciphered a holistic overview of cancer behaviors, yielding precise molecular classification to facilitate the evolution and refinement of precision medicine. CONCLUSION This review outlined the biomarkers at multiple expression layers to tutor molecular classification and pinpoint tumor diagnosis, and explored the paradigm shift in precision therapy: from single- to multi-omics-based subtyping to optimize therapeutic regimens. Ultimately, we firmly believe that by parsing molecular characteristics, omics-based typing will be a powerful assistant for precision oncology.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ting Lin
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Haijiao Zou
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Aoyang Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
47
|
Lynch SM, Heeran AB, Burke C, Lynam-Lennon N, Eustace AJ, Dean K, Robson T, Rahman A, Marcone S. Precision Oncology, Artificial Intelligence, and Novel Therapeutic Advancements in the Diagnosis, Prevention, and Treatment of Cancer: Highlights from the 59th Irish Association for Cancer Research (IACR) Annual Conference. Cancers (Basel) 2024; 16:1989. [PMID: 38893110 PMCID: PMC11171401 DOI: 10.3390/cancers16111989] [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: 04/19/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Advancements in oncology, especially with the era of precision oncology, is resulting in a paradigm shift in cancer care. Indeed, innovative technologies, such as artificial intelligence, are paving the way towards enhanced diagnosis, prevention, and personalised treatments as well as novel drug discoveries. Despite excellent progress, the emergence of resistant cancers has curtailed both the pace and extent to which we can advance. By combining both their understanding of the fundamental biological mechanisms and technological advancements such as artificial intelligence and data science, cancer researchers are now beginning to address this. Together, this will revolutionise cancer care, by enhancing molecular interventions that may aid cancer prevention, inform clinical decision making, and accelerate the development of novel therapeutic drugs. Here, we will discuss the advances and approaches in both artificial intelligence and precision oncology, presented at the 59th Irish Association for Cancer Research annual conference.
Collapse
Affiliation(s)
- Seodhna M. Lynch
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, Londonderry BT47 6SB, UK;
| | - Aisling B. Heeran
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St. James’s Cancer Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland; (A.B.H.); (N.L.-L.); (S.M.)
| | - Caoimbhe Burke
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, D04 C1P1 Dublin, Ireland;
| | - Niamh Lynam-Lennon
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St. James’s Cancer Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland; (A.B.H.); (N.L.-L.); (S.M.)
| | - Alex J. Eustace
- Life Sciences Institute, Dublin City University, D09 NR58 Dublin, Ireland;
| | - Kellie Dean
- School of Biochemistry and Cell Biology, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland;
| | - Tracy Robson
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Arman Rahman
- UCD School of Medicine, UCD Conway Institute, University College Dublin, Belfield, D04 C1P1 Dublin, Ireland;
| | - Simone Marcone
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St. James’s Cancer Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland; (A.B.H.); (N.L.-L.); (S.M.)
| |
Collapse
|
48
|
Fu Y, Wang C, Wu Z, Zhang X, Liu Y, Wang X, Liu F, Chen Y, Zhang Y, Zhao H, Wang Q. Discovery of the potential biomarkers for early diagnosis of endometrial cancer via integrating metabolomics and transcriptomics. Comput Biol Med 2024; 173:108327. [PMID: 38552279 DOI: 10.1016/j.compbiomed.2024.108327] [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: 02/06/2024] [Revised: 03/07/2024] [Accepted: 03/17/2024] [Indexed: 04/17/2024]
Abstract
Endometrial cancer (EC) is one of the most common malignant tumors in women, and the increasing incidence and mortality pose a serious threat to the public health. Early diagnosis of EC could prolong the survival period and optimize the survivorship, greatly alleviating patients' suffering and social medical pressure. In this study, we collected urine and serum samples from the recruited patients, analyzed the samples using LC-MS approach, and identified the differential metabolites through metabolomic analysis. Then, the differentially expressed genes were identified through the systematic transcriptomic analysis of EC-related dataset from Gene Expression Omnibus (GEO), followed by network profiling of metabolic-reaction-enzyme-gene. In this experiment, a total of 83 differential metabolites and 19 hub genes were discovered, of which 10 different metabolites and 3 hub genes were further evaluated as more potential biomarkers based on network analysis. According to the KEGG enrichment analysis, the potential biomarkers and gene-encoded proteins were found to be involved in the arginine and proline metabolism, histidine metabolism, and pyrimidine metabolism, which was of significance for the early diagnosis of EC. In particular, the combination of metabolites (histamine, 1-methylhistamine, and methylimidazole acetaldehyde) as well as the combination of RRM2, TYMS and TK1 exerted more accurate discrimination abilities between EC and healthy groups, providing more criteria for the early diagnosis of EC.
Collapse
Affiliation(s)
- Yan Fu
- School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China; Core Facilities and Centers, Hebei Medical University, Shijiazhuang, 050017, China
| | - Chengzhao Wang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zhimin Wu
- School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xiaoguang Zhang
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, 050017, China; College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yan Liu
- School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xu Wang
- School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Fangfang Liu
- School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yujuan Chen
- School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yang Zhang
- School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Huanhuan Zhao
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, 050011, China.
| | - Qiao Wang
- School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
49
|
Ramachandran A, Dhar R, Devi A. Stem Cell-Derived Exosomes: An Advanced Horizon to Cancer Regenerative Medicine. ACS APPLIED BIO MATERIALS 2024; 7:2128-2139. [PMID: 38568170 DOI: 10.1021/acsabm.4c00089] [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] [Indexed: 04/16/2024]
Abstract
Cancer research has made significant progress in recent years, and extracellular vesicles (EVs) based cancer investigation reveals several facts about cancer. Exosomes are a subpopulation of EVs. In the present decade, exosomes is mostly highlighted for cancer theranostic research. Tumor cell derived exosomes (TEXs) promote cancer but there are multiple sources of exosomes that can be used as cancer therapeutic agents (plant exosomes, stem cell-derived exosomes, modified or synthetic exosomes). Stem cells based regenerative medicine faces numerous challenges, such as promote tumor development, cellular reprogramming etc., and therefore addressing these complications becomes essential. Stem cell-derived exosomes serves as an answer to these problems and offers a better solution. Global research indicates that stem cell-derived exosomes also play a dual role in the cellular system by either inhibiting or promoting cancer. Modified exosomes which are genetically engineered exosomes or surface modified exosomes to increase the efficacy of the therapeutic properties can also be considered to target the above concerns. However, the difficulties associated with the exosomes include variations in exosomes heterogenity, isolation protocols, large scale production, etc., and these have to be managed effectively. In this review, we explore exosomes biogenesis, multiple stem cell-derived exosome sources, drug delivery, modified stem cells exosomes, clinical trial of stem cells exosomes, and the related challenges in this domain and future orientation. This article may encourage researchers to explore stem cell-derived exosomes and develop an effective and affordable cancer therapeutic solution.
Collapse
Affiliation(s)
- Aparna Ramachandran
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| |
Collapse
|
50
|
Chitluri KK, Emerson IA. The importance of protein domain mutations in cancer therapy. Heliyon 2024; 10:e27655. [PMID: 38509890 PMCID: PMC10950675 DOI: 10.1016/j.heliyon.2024.e27655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 03/22/2024] Open
Abstract
Cancer is a complex disease that is caused by multiple genetic factors. Researchers have been studying protein domain mutations to understand how they affect the progression and treatment of cancer. These mutations can significantly impact the development and spread of cancer by changing the protein structure, function, and signalling pathways. As a result, there is a growing interest in how these mutations can be used as prognostic indicators for cancer prognosis. Recent studies have shown that protein domain mutations can provide valuable information about the severity of the disease and the patient's response to treatment. They may also be used to predict the response and resistance to targeted therapy in cancer treatment. The clinical implications of protein domain mutations in cancer are significant, and they are regarded as essential biomarkers in oncology. However, additional techniques and approaches are required to characterize changes in protein domains and predict their functional effects. Machine learning and other computational tools offer promising solutions to this challenge, enabling the prediction of the impact of mutations on protein structure and function. Such predictions can aid in the clinical interpretation of genetic information. Furthermore, the development of genome editing tools like CRISPR/Cas9 has made it possible to validate the functional significance of mutants more efficiently and accurately. In conclusion, protein domain mutations hold great promise as prognostic and predictive biomarkers in cancer. Overall, considerable research is still needed to better define genetic and molecular heterogeneity and to resolve the challenges that remain, so that their full potential can be realized.
Collapse
Affiliation(s)
- Kiran Kumar Chitluri
- Bioinformatics Programming Lab, Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, TN, 632014, India
| | - Isaac Arnold Emerson
- Bioinformatics Programming Lab, Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, TN, 632014, India
| |
Collapse
|