1
|
Hsu W, Prosper AE. The Need for Contemporary, Diverse, and Annotated Datasets to Advance AI in Lung Cancer Screening. Radiol Artif Intell 2025; 7:e250371. [PMID: 40497818 DOI: 10.1148/ryai.250371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2025]
Affiliation(s)
- William Hsu
- Department of Radiological Sciences, University of California Los Angeles, 924 Westwood Blvd, Ste 420, Los Angeles, CA 90095
| | - Ashley E Prosper
- Department of Radiological Sciences, University of California Los Angeles, 924 Westwood Blvd, Ste 420, Los Angeles, CA 90095
| |
Collapse
|
2
|
Xu J, Wang T, Li J, Wang Y, Zhu Z, Fu X, Wang J, Zhang Z, Cai W, Song R, Hou C, Yang LZ, Wang H, Wong STC, Li H. A multimodal fusion system predicting survival benefits of immune checkpoint inhibitors in unresectable hepatocellular carcinoma. NPJ Precis Oncol 2025; 9:185. [PMID: 40517171 PMCID: PMC12167364 DOI: 10.1038/s41698-025-00979-6] [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: 12/31/2024] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
Early identification of unresectable hepatocellular carcinoma (HCC) patients who may benefit from immune checkpoint inhibitors (ICIs) is crucial for optimizing outcomes. Here, we developed a multimodal fusion (MMF) system integrating CT-derived deep learning features and clinical data to predict overall survival (OS) and progression-free survival (PFS). Using retrospective multicenter data (n = 859), the MMF combining an ensemble deep learning (Ensemble-DL) model with clinical variables achieved strong external validation performance (C-index: OS = 0.74, PFS = 0.69), outperforming radiomics (29.8% OS improvement), mRECIST (27.6% OS improvement), clinical benchmarks (C-index: OS = 0.67, p = 0.0011; PFS = 0.65, p = 0.033), and Ensemble-DL (C-index: OS = 0.69, p = 0.0028; PFS = 0.66, p = 0.044). The MMF system effectively stratified patients across clinical subgroups and demonstrated interpretability through activation maps and radiomic correlations. Differential gene expression analysis revealed enrichment of the PI3K/Akt pathway in patients identified by the MMF system. The MMF system provides an interpretable, clinically applicable approach to guide personalized ICI treatment in unresectable HCC.
Collapse
Affiliation(s)
- Jun Xu
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
- University of Science and Technology of China, Hefei, P. R. China
- Department of Oncology, Hefei Cancer Hospital; Chinese Academy of Sciences, Hefei, P. R. China
- Department of Interventional Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
| | - Tengfei Wang
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
- University of Science and Technology of China, Hefei, P. R. China
- Department of Oncology, Hefei Cancer Hospital; Chinese Academy of Sciences, Hefei, P. R. China
| | - Junjun Li
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, P. R. China
| | - Yong Wang
- Department of Radiology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, P. R. China
| | - Zhangxiang Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, P. R. China
| | - Xiao Fu
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
- Department of Oncology, Hefei Cancer Hospital; Chinese Academy of Sciences, Hefei, P. R. China
| | - Junjie Wang
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
- University of Science and Technology of China, Hefei, P. R. China
| | - Zhenglin Zhang
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
- University of Science and Technology of China, Hefei, P. R. China
| | - Wei Cai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, the University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, P. R. China
| | - Ruipeng Song
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, the University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, P. R. China
| | - Changlong Hou
- Department of Interventional Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
| | - Li-Zhuang Yang
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
- University of Science and Technology of China, Hefei, P. R. China
- Department of Oncology, Hefei Cancer Hospital; Chinese Academy of Sciences, Hefei, P. R. China
| | - Hongzhi Wang
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
- University of Science and Technology of China, Hefei, P. R. China
- Department of Oncology, Hefei Cancer Hospital; Chinese Academy of Sciences, Hefei, P. R. China
| | - Stephen T C Wong
- Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, TX, USA.
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA.
| | - Hai Li
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China.
- University of Science and Technology of China, Hefei, P. R. China.
- Department of Oncology, Hefei Cancer Hospital; Chinese Academy of Sciences, Hefei, P. R. China.
| |
Collapse
|
3
|
Wei Z, Yu R, Zhang Y, Wang Y, Wang J, Xie C, Chen X. A CT-based radiomic model for predicting vertebral fractures in older patients with type 2 diabetes mellitus: A longitudinal study. J Endocrinol Invest 2025:10.1007/s40618-025-02627-z. [PMID: 40493166 DOI: 10.1007/s40618-025-02627-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Accepted: 06/05/2025] [Indexed: 06/12/2025]
Affiliation(s)
- Zicheng Wei
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Rui Yu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yiping Zhang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yu Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Jianhua Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Cao Xie
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Xiao Chen
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
4
|
Hsu CX, Chou HC, Yen SH. Exploring emergency presentations management in lung cancer: insights on oncology emergencies and histological heterogeneity. Am J Emerg Med 2025; 92:198-199. [PMID: 39730277 DOI: 10.1016/j.ajem.2024.12.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/29/2024] Open
Affiliation(s)
- Chen-Xiong Hsu
- Division of Radiation Oncology and Hyperthermia Center, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Department of Computer Science and Engineering, Yuan Ze University, Taoyuan, Taiwan.
| | - Hui-Chen Chou
- Division of Radiation Oncology and Hyperthermia Center, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Department of Nursing, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Sang-Hue Yen
- Department of Radiation Oncology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
5
|
Su Y, Tao J, Lan X, Liang C, Huang X, Zhang J, Li K, Chen L. CT-based intratumoral and peritumoral radiomics nomogram to predict spread through air spaces in lung adenocarcinoma with diameter ≤ 3 cm: A multicenter study. Eur J Radiol Open 2025; 14:100630. [PMID: 39850145 PMCID: PMC11754163 DOI: 10.1016/j.ejro.2024.100630] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025] Open
Abstract
Purpose The aim of this study was to explore and develop a preoperative and noninvasive model for predicting spread through air spaces (STAS) status in lung adenocarcinoma (LUAD) with diameter ≤ 3 cm. Methods This multicenter retrospective study included 640 LUAD patients. Center I included 525 patients (368 in the training cohort and 157 in the validation cohort); center II included 115 patients (the test cohort). We extracted radiomics features from the intratumor, extended tumor and peritumor regions. Multivariate logistic regression and boruta algorithm were used to select clinical independent risk factors and radiomics features, respectively. We developed a clinical model and four radiomics models (the intratumor model, extended tumor model, peritumor model and fusion model). A nomogram based on prediction probability value of the optimal radiomics model and clinical independent risk factors was developed to predict STAS status. Results Maximum diameter and nodule type were clinical independent risk factors. The extended tumor model achieved satisfactory STAS status discrimination performance with the AUC of 0.74, 0.71 and 0.80 in the three cohorts, respectively, performed better than other radiomics models. The integrated discrimination improvement value revealed that the nomogram outperformed compared to the clinical model with the value of 12 %. Patients with high nomogram score (≥ 77.31) will be identified as STAS-positive. Conclusions Peritumoral information is significant to predict STAS status. The nomogram based on the extended tumor model and clinical independent risk factors provided good preoperative prediction of STAS status in LUAD with diameter ≤ 3 cm, aiding surgical decision-making.
Collapse
Affiliation(s)
- Yangfan Su
- Department of Radiology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
| | - Junli Tao
- Department of Radiology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
| | - Xiaosong Lan
- Department of Radiology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
| | - Changyu Liang
- Department of Radiology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
| | - Xuemei Huang
- Department of Radiology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
| | - Jiuquan Zhang
- Department of Radiology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
| | - Kai Li
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong road, Qingxiu district, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Lihua Chen
- Department of Radiology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu road, Shapingba district, Chongqing 400030, China
| |
Collapse
|
6
|
Alizade-Harakiyan M, Khodaei A, Yousefi A, Zamani H, Mesbahi A. Decision tree-based machine learning algorithm for prediction of acute radiation esophagitis. Biochem Biophys Rep 2025; 42:101991. [PMID: 40230494 PMCID: PMC11995095 DOI: 10.1016/j.bbrep.2025.101991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
Background Radiation-induced esophagitis remains a significant challenge in thoracic and neck cancer treatment, impacting patient quality of life and potentially limiting therapeutic efficacy. This study aimed to develop and validate a decision tree-based model for predicting acute esophagitis grades in patients undergoing chemoradiotherapy. Methods Data from 100 patients receiving thoracic and neck radiotherapy were analyzed. The dataset comprised 33 features, including demographic, clinical, and dosimetric parameters. A decision tree classifier was implemented for both binary (Grade ≥2 vs. <2) and multi-class (Grades 1, 2, and 3) classification. Model performance was evaluated using standard metrics including accuracy, precision, recall, and F1-score. Results The binary classification model achieved 97 % accuracy in distinguishing acute esophagitis. The multi-class model demonstrated 98 % accuracy in predicting specific grades. Key predictive features included V40 (volume receiving 40 Gy), V60, and average esophageal dose. The model generated interpretable decision rules, with V60 ≥ 2.3 strongly indicating Grade 3 esophagitis. Conclusions The decision tree model demonstrates high accuracy in predicting radiation-induced esophagitis grades while maintaining clinical interpretability. This approach offers potential for treatment optimization and personalized risk assessment in radiotherapy planning. The model's transparency and reliability make it a promising tool for clinical decision support in radiation oncology.
Collapse
Affiliation(s)
- Mostafa Alizade-Harakiyan
- Department of Radiation Oncology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Khodaei
- Faculty of Electrical and Computer Engineering, University of Tabriz, Tabriz, Iran
| | - Ali Yousefi
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Zamani
- Medical Physics Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asghar Mesbahi
- Medical Physics Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
Shoaib M, Tariq A, Liu Y, Yang M, Qu L, Yang L, Song J. Recent update on the development of HPV16 inhibitors for cervical cancer. Crit Rev Oncol Hematol 2025; 210:104703. [PMID: 40107437 DOI: 10.1016/j.critrevonc.2025.104703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
Persistent infection with human papillomavirus (HPV) can lead to cervical cancer (CC), which is the fourth most commonly diagnosed cancer in women globally. In this review, we have explained the HPV genome and the development of CC. Additionally, we summarized recently discovered small molecules that act as inhibitors of HPV-16. These molecules were identified through experimental and in-silico studies aimed at preventing or treating CC. HPV-16 and HPV-18 are the most common subtypes of HPV that cause CC globally. E6 oncoprotein of HPV-16 is considered the primary cause of CC progression. Therefore, E6 is the most focused targeted protein for developing specific and novel therapeutic inhibitors to treat HPV-related cancers. In recent years, various HPV inhibitors have been identified by means of experimental and in-silico studies. In addition, artificial intelligence-based medical diagnostic tools have grown more popular as they are capable of screening and diagnosing HPV-related cancer.
Collapse
Affiliation(s)
- Muhammad Shoaib
- College of Chemistry, Pingyuan Laboratory, and State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Amina Tariq
- College of Chemistry, Pingyuan Laboratory, and State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yanchen Liu
- College of Chemistry, Pingyuan Laboratory, and State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Mingwei Yang
- College of Chemistry, Pingyuan Laboratory, and State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Lingbo Qu
- College of Chemistry, Pingyuan Laboratory, and State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Zhengzhou University, Zhengzhou, Henan 450001, China; Institute of Chemistry, Henan Academy of Science, Zhengzhou, Henan 450046, China
| | - Longhua Yang
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jinshuai Song
- College of Chemistry, Pingyuan Laboratory, and State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
8
|
Vyas A, Kumar K, Sharma A, Verma D, Bhatia D, Wahi N, Yadav AK. Advancing the frontier of artificial intelligence on emerging technologies to redefine cancer diagnosis and care. Comput Biol Med 2025; 191:110178. [PMID: 40228444 DOI: 10.1016/j.compbiomed.2025.110178] [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: 01/30/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Artificial Intelligence (AI) is capable of revolutionizing cancer therapy and advancing precision oncology via integrating genomics data and digitized health information. AI applications show promise in cancer prediction, prognosis, and treatment planning, particularly in radiomics, deep learning, and machine learning for early cancer diagnosis. However, widespread adoption requires comprehensive data and clinical validation. While AI has demonstrated advantages in treating common malignancies like lung and breast cancers, challenges remain in managing rare tumors due to limited datasets. AI's role in processing multi-omics data and supporting precision oncology decision-making is critical as genetic and health data become increasingly digitized. METHOD This review article presents current knowledge on AI and associated technologies, which are being utilized in the diagnosis and therapy of cancer. The applications of AI in radiomics, deep learning, and machine learning for cancer screening and treatment planning are examined. The study also explores the capabilities and limitations of predictive AI in diagnosis and prognosis, as well as generative AI, such as advanced chatbots, in patient and provider interactions. RESULTS AI can improve the early diagnosis and treatment of high-incidence cancers like breast and lung cancer. However, its application in rare cancers is limited by insufficient data for training and validation. AI can effectively process large-scale multi-omics data from DNA and RNA sequencing, enhancing precision oncology. Predictive AI aids in risk assessment and prognosis, while generative AI tools improve patient-provider communication. Despite these advancements, further research and technological progress are needed to overcome existing challenges. CONCLUSIONS AI holds transformative potential for cancer therapy, particularly in precision oncology, early detection, and personalized treatment planning. However, challenges such as data limitations in rare cancers, the need for clinical validation, and regulatory considerations must be addressed. Future advancements in AI could significantly improve decision-support systems in oncology, ultimately enhancing patient care and quality of life. The review highlights both the opportunities and obstacles in integrating AI into cancer diagnostics and therapeutics, calling for continued research and regulatory oversight.
Collapse
Affiliation(s)
- Akanksha Vyas
- Academy of Scientific and Innovative Research, Ghaziabad, 201002, India
| | - Krishan Kumar
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Ayushi Sharma
- College of Medicine, Taipei Medical University, Taipei City, 110, Taiwan
| | - Damini Verma
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Dhiraj Bhatia
- Department of Biological Sciences & Engineering, Indian Institute of Technology Gandhinagar, Near Palaj, Gandhinagar, Gujarat, 382355, India
| | - Nitin Wahi
- Department of Biotechnology, LNCT University, Kolar Road, Shirdipuram, Bhopal, Madhya Pradesh, 462042, India
| | - Amit K Yadav
- Department of Biological Sciences & Engineering, Indian Institute of Technology Gandhinagar, Near Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
9
|
Hassan M, Shahzadi S, Kloczkowski A. Harnessing Artificial Intelligence in Pediatric Oncology Diagnosis and Treatment: A Review. Cancers (Basel) 2025; 17:1828. [PMID: 40507308 PMCID: PMC12153614 DOI: 10.3390/cancers17111828] [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: 04/22/2025] [Revised: 05/25/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025] Open
Abstract
Artificial intelligence (AI) is rapidly transforming pediatric oncology by creating new means to improve the accuracy and efficacy of cancer diagnosis and treatment in children. This review critically examines current applications of AI technologies like machine learning (ML) and deep learning (DL) to the main types of pediatric cancers. However, the application of AI to pediatric oncology is prone to certain challenges, including the heterogeneity and rarity of pediatric cancer data, rapid technological development in imaging, and ethical concerns pertaining to data privacy and algorithmic transparency. Collaborative efforts and data-sharing schemes are important to surpass these challenges and facilitate effective training of AI models. This review also points to emerging trends, including AI-based radiomics and proteomics applications, and provides future directions to realize the full potential of AI in pediatric oncology. Finally, AI is a promising paradigm shift toward precision medicine in childhood cancer treatment, which can enhance the survival rates and quality of life for pediatric patients.
Collapse
Affiliation(s)
- Mubashir Hassan
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (M.H.); (S.S.)
| | - Saba Shahzadi
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (M.H.); (S.S.)
| | - Andrzej Kloczkowski
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (M.H.); (S.S.)
- Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
10
|
Kumar R, Sporn K, Khanna A, Paladugu P, Gowda C, Ngo A, Jagadeesan R, Zaman N, Tavakkoli A. Integrating Radiogenomics and Machine Learning in Musculoskeletal Oncology Care. Diagnostics (Basel) 2025; 15:1377. [PMID: 40506947 PMCID: PMC12155258 DOI: 10.3390/diagnostics15111377] [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: 05/04/2025] [Revised: 05/21/2025] [Accepted: 05/23/2025] [Indexed: 06/16/2025] Open
Abstract
Musculoskeletal tumors present a diagnostic challenge due to their rarity, histological diversity, and overlapping imaging features. Accurate characterization is essential for effective treatment planning and prognosis, yet current diagnostic workflows rely heavily on invasive biopsy and subjective radiologic interpretation. This review explores the evolving role of radiogenomics and machine learning in improving diagnostic accuracy for bone and soft tissue tumors. We examine integrating quantitative imaging features from MRI, CT, and PET with genomic and transcriptomic data to enable non-invasive tumor profiling. AI-powered platforms employing convolutional neural networks (CNNs) and radiomic texture analysis show promising results in tumor grading, subtype differentiation (e.g., Osteosarcoma vs. Ewing sarcoma), and predicting mutation signatures (e.g., TP53, RB1). Moreover, we highlight the use of liquid biopsy and circulating tumor DNA (ctDNA) as emerging diagnostic biomarkers, coupled with point-of-care molecular assays, to enable early and accurate detection in low-resource settings. The review concludes by discussing translational barriers, including data harmonization, regulatory challenges, and the need for multi-institutional datasets to validate AI-based diagnostic frameworks. This article synthesizes current advancements and provides a forward-looking view of precision diagnostics in musculoskeletal oncology.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.G.); (A.N.)
| | - Kyle Sporn
- Norton College of Medicine, Upstate Medical University, Syracuse, NY 13210, USA;
| | - Akshay Khanna
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.K.); (P.P.)
| | - Phani Paladugu
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.K.); (P.P.)
- Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Chirag Gowda
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.G.); (A.N.)
| | - Alex Ngo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.G.); (A.N.)
| | - Ram Jagadeesan
- Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA;
- Cisco AI Systems, Cisco Inc., San Jose, CA 95134, USA
| | - Nasif Zaman
- Department of Computer Science, University of Nevada Reno, Reno, NV 89557, USA; (N.Z.); (A.T.)
| | - Alireza Tavakkoli
- Department of Computer Science, University of Nevada Reno, Reno, NV 89557, USA; (N.Z.); (A.T.)
| |
Collapse
|
11
|
Amante E, Ghyselinck R, Thiberville L, Trisolini R, Guisier F, Delchevalerie V, Dumas B, Frénay B, Duparc I, Mazellier N, Farhi C, Jubert C, Salaün M, Lachkar S. Human and Deep Learning Predictions of Peripheral Lung Cancer Using a 1.3 mm Video Endoscopic Probe. Respirology 2025. [PMID: 40433758 DOI: 10.1111/resp.70057] [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: 11/12/2024] [Revised: 02/16/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND AND OBJECTIVE Iriscope, a 1.3 mm video endoscopic probe introduced through an r-EBUS catheter, allows for the direct visualisation of small peripheral pulmonary nodules (PPNs). This study assessed the ability of physicians with different levels of experience in bronchoscopy, and the ability of artificial intelligence (AI) to predict the malignant nature of small PPNs during Iriscope peripheral endoscopy. METHODS Patients undergoing bronchoscopy with r-EBUS and Iriscope for peripheral PPNs < 20 mm with a definite diagnosis were analysed. Senior and Junior physicians independently interpreted video-recorded Iriscope sequences, classifying them as tumoral (malignant) or non-tumoral, blind to the final diagnosis. A deep learning (DL) model was also trained on Iriscope images and tested on a different set of patients for comparison with human interpretation. Diagnostic accuracy, sensitivity, specificity, and F1 score were calculated. RESULTS Sixty-one patients with small PPNs (median size 15 mm, IQR: 11-20 mm) were included. The technique allowed for the direct visualisation of the lesions in all cases. The final diagnosis was cancer for 37 cases and a benign lesion in 24 cases. Senior physicians outperformed junior physicians in recognising tumoral Iriscope images, with a balanced accuracy of 85.4% versus 66.7%, respectively, when compared with the final diagnosis. The DL model outperformed junior physicians with a balanced accuracy of 71.5% but was not superior to senior physicians. CONCLUSION Iriscope could be a valuable tool in PPNs management, especially for experienced operators. Applied to Iriscope images, DL could enhance overall performance of less experienced physicians in diagnosing malignancy.
Collapse
Affiliation(s)
- Edoardo Amante
- Department of Pneumology, Rouen University Hospital, Rouen, France
- Department of Pulmonary Medicine and Interventional Pulmonology, Catholic University of the Sacred Hearth, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Robin Ghyselinck
- Faculty of Computer Science, NaDI, University of Namur, Namur, Belgium
| | - Luc Thiberville
- Department of Pneumology, Rouen University Hospital, Rouen, France
| | - Rocco Trisolini
- Department of Pulmonary Medicine and Interventional Pulmonology, Catholic University of the Sacred Hearth, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Florian Guisier
- Department of Pneumology, Rouen University Hospital, Rouen, France
- Department of Pulmonology and Inserm CIC-CRB 1404, Normandie Univ, UNIROUEN, LITIS Lab QuantIF Team EA4108, CHU Rouen, Rouen, France
| | | | - Bruno Dumas
- Faculty of Computer Science, NaDI, University of Namur, Namur, Belgium
| | - Benoît Frénay
- Faculty of Computer Science, NaDI, University of Namur, Namur, Belgium
| | - Inès Duparc
- Department of Pneumology, Rouen University Hospital, Rouen, France
| | | | - Cecile Farhi
- Department of Pneumology, Rouen University Hospital, Rouen, France
| | | | - Mathieu Salaün
- Department of Pneumology, Rouen University Hospital, Rouen, France
- Department of Pulmonology and Inserm CIC-CRB 1404, Normandie Univ, UNIROUEN, LITIS Lab QuantIF Team EA4108, CHU Rouen, Rouen, France
| | - Samy Lachkar
- Department of Pneumology, Rouen University Hospital, Rouen, France
- Department of Pulmonology and Inserm CIC-CRB 1404, Normandie Univ, UNIROUEN, LITIS Lab QuantIF Team EA4108, CHU Rouen, Rouen, France
| |
Collapse
|
12
|
Zhang R, Zhang X, Dou Z, Lin J, Qin S, Xu C, Chen Y, Zhu J, Wang J. Machine learning-based radiomics analysis in enhancing CT for predicting pathological subtypes and WHO staging of thymic epithelial tumors: a multicenter study. Am J Cancer Res 2025; 15:2375-2396. [PMID: 40520864 PMCID: PMC12163439 DOI: 10.62347/stuz8659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/14/2025] [Indexed: 06/18/2025] Open
Abstract
This study is aimed to develop predictive models for classifying thymic epithelial tumor (TET) histological subtypes (A/AB/B1, B2/B3, C) and WHO stages (I-IV) using radiomics features derived from contrast-enhanced CT scans. These models were validated on multicenter external datasets to improve preoperative diagnosis and guide treatment decisions. A total of 257 patients diagnosed with TET between January 2013 and April 2024 were retrospectively analyzed, with 181 cases from the First Affiliated Hospital of Soochow University served as the training cohort and 76 cases from the Second Affiliated Hospital used as an external test set. All patients underwent preoperative enhanced CT scans. After manual segmentation of the volume of interest (VOI), 1,038 radiomic features were extracted. Feature selection was performed using PCA and LASSO methods. Three models (clinical semantic, radiomics, and a fusion model combining both) were built using random forest algorithms. The fusion model achieved the highest performance in the external test set, with an accuracy of 0.908 and F1 score of 0.896 for histological subtype classification, and an accuracy of 0.803 and F1 score of 0.833 for WHO staging. The radiomics model shows slightly lower performance, while the clinical semantic model performs the weakest. Our findings suggest that machine learning models integrating radiomics and clinical features can effectively predict TET subtypes and stages, offering a non-invasive tool for accurate preoperative assessment with strong generalization ability.
Collapse
Affiliation(s)
- Ruoxu Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Xueyi Zhang
- Department of General Surgery, Changshu Hospital Affiliated to Soochow UniversitySuzhou, Jiangsu, China
| | - Zheng Dou
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Jiaxi Lin
- Department of Gastroenterology, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Songbing Qin
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Chao Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Yongbing Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Jinzhou Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Jianping Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| |
Collapse
|
13
|
Akbari H, Bakas S, Sako C, Fathi Kazerooni A, Villanueva-Meyer J, Garcia JA, Mamourian E, Liu F, Cao Q, Shinohara RT, Baid U, Getka A, Pati S, Singh A, Calabrese E, Chang S, Rudie J, Sotiras A, LaMontagne P, Marcus DS, Milchenko M, Nazeri A, Balana C, Capellades J, Puig J, Badve C, Barnholtz-Sloan JS, Sloan AE, Vadmal V, Waite K, Ak M, Colen RR, Park YW, Ahn SS, Chang JH, Choi YS, Lee SK, Alexander GS, Ali AS, Dicker AP, Flanders AE, Liem S, Lombardo J, Shi W, Shukla G, Griffith B, Poisson LM, Rogers LR, Kotrotsou A, Booth TC, Jain R, Lee M, Mahajan A, Chakravarti A, Palmer JD, DiCostanzo D, Fathallah-Shaykh H, Cepeda S, Santonocito OS, Di Stefano AL, Wiestler B, Melhem ER, Woodworth GF, Tiwari P, Valdes P, Matsumoto Y, Otani Y, Imoto R, Aboian M, Koizumi S, Kurozumi K, Kawakatsu T, Alexander K, Satgunaseelan L, Rulseh AM, Bagley SJ, Bilello M, Binder ZA, Brem S, Desai AS, Lustig RA, Maloney E, Prior T, Amankulor N, Nasrallah MP, O’Rourke DM, Mohan S, Davatzikos C. Machine learning-based prognostic subgrouping of glioblastoma: A multicenter study. Neuro Oncol 2025; 27:1102-1115. [PMID: 39665363 PMCID: PMC12083074 DOI: 10.1093/neuonc/noae260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most aggressive adult primary brain cancer, characterized by significant heterogeneity, posing challenges for patient management, treatment planning, and clinical trial stratification. METHODS We developed a highly reproducible, personalized prognostication, and clinical subgrouping system using machine learning (ML) on routine clinical data, magnetic resonance imaging (MRI), and molecular measures from 2838 demographically diverse patients across 22 institutions and 3 continents. Patients were stratified into favorable, intermediate, and poor prognostic subgroups (I, II, and III) using Kaplan-Meier analysis (Cox proportional model and hazard ratios [HR]). RESULTS The ML model stratified patients into distinct prognostic subgroups with HRs between subgroups I-II and I-III of 1.62 (95% CI: 1.43-1.84, P < .001) and 3.48 (95% CI: 2.94-4.11, P < .001), respectively. Analysis of imaging features revealed several tumor properties contributing unique prognostic value, supporting the feasibility of a generalizable prognostic classification system in a diverse cohort. CONCLUSIONS Our ML model demonstrates extensive reproducibility and online accessibility, utilizing routine imaging data rather than complex imaging protocols. This platform offers a unique approach to personalized patient management and clinical trial stratification in GBM.
Collapse
Affiliation(s)
- Hamed Akbari
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, California, USA
| | - Spyridon Bakas
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Computer Science, Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, Indiana, USA
| | - Chiharu Sako
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anahita Fathi Kazerooni
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Data-Driven Discovery in Biomedicine (D3b), Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Javier Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Jose A Garcia
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth Mamourian
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fang Liu
- Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Quy Cao
- Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Russell T Shinohara
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ujjwal Baid
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Alexander Getka
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sarthak Pati
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ashish Singh
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Evan Calabrese
- Department of Radiology, Duke University, Durham, North Carolina, USA
| | - Susan Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey Rudie
- Department of Radiology, University of California San Diego, San Diego, California, USA
| | - Aristeidis Sotiras
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mikhail Milchenko
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Arash Nazeri
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Carmen Balana
- B-ARGO Group, Institut Investigació Germans Trias i Pujol (IGTP), Badalona (Barcelona), Catalonia, Spain
| | - Jaume Capellades
- Research Unit (IDIR), Image Diagnosis Institute, Badalona, Spain
| | - Josep Puig
- Department of Radiology (CDI), Hospital Clínic and IDIBAPS, Barcelona, Spain
| | - Chaitra Badve
- Department of Radiology, Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| | - Jill S Barnholtz-Sloan
- Trans-Divisional Research Program (TDRP), Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute, Bethesda, Maryland, USA
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois, USA
- Center for Biomedical Informatics and Information Technology (CBIIT), National Cancer Institute, Bethesda, Maryland, USA
| | - Andrew E Sloan
- Brain and Tumor Neurosurgery, Neurosurgical Oncology, Piedmont Health, Atlanta, Georgia, USA
- Seidman Cancer Center and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Vachan Vadmal
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Kristin Waite
- Center for Biomedical Informatics and Information Technology (CBIIT), National Cancer Institute, Bethesda, Maryland, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Murat Ak
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rivka R Colen
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Yae Won Park
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung Soo Ahn
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Tumor Center, Severance Hospital, Yonsei University Health System, Seoul, Republic of Korea
| | - Yoon Seong Choi
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Seung-Koo Lee
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gregory S Alexander
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ayesha S Ali
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adam P Dicker
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adam E Flanders
- Department of Radiology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Spencer Liem
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Joseph Lombardo
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Wenyin Shi
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Gaurav Shukla
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Radiation Oncology, Christiana Care Health System, Philadelphia, Pennsylvania, USA
| | - Brent Griffith
- Department of Radiology, Henry Ford Health System, Detroit, Michigan, USA
| | - Laila M Poisson
- Department of Public Health Sciences, Center for Bioinformatics, Henry Ford Health System, Detroit, Michigan, USA
- Department of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Cancer Institute, Henry Ford Health, Detroit, Michigan, USA
| | - Lisa R Rogers
- Department of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Cancer Institute, Henry Ford Health, Detroit, Michigan, USA
| | | | - Thomas C Booth
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
- Department of Neuroradiology, Ruskin Wing, King’s College Hospital NHS Foundation Trust, London, UK
| | - Rajan Jain
- Department of Radiology, New York University Langone Health, New York, New York, USA
- Department of Neurosurgery, New York University Langone Health, New York, New York, USA
| | - Matthew Lee
- Department of Radiology, New York University Langone Health, New York, New York, USA
| | - Abhishek Mahajan
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK
| | - Arnab Chakravarti
- Department of Radiation Oncology, The James Cancer Hospital at the Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Joshua D Palmer
- Department of Radiation Oncology, The James Cancer Hospital at the Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Dominic DiCostanzo
- Department of Radiation Oncology, The James Cancer Hospital at the Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | | | - Santiago Cepeda
- Department of Neurosurgery, University Hospital Río Hortega, Valladolid, Spain
| | - Orazio Santo Santonocito
- Division of Neurosurgery, Spedali Riuniti di Livorno-Azienda USL Toscana Nord-Ovest, Livorno, Italy
| | - Anna Luisa Di Stefano
- Division of Neurosurgery, Spedali Riuniti di Livorno-Azienda USL Toscana Nord-Ovest, Livorno, Italy
| | - Benedikt Wiestler
- Department of Neuroradiology, Technical University of Munich, Munchen, Germany
| | - Elias R Melhem
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Graeme F Woodworth
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Pallavi Tiwari
- Department of Radiology, University of Wisconsin, Madison, Wisconsin, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, USA
| | - Pablo Valdes
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yuji Matsumoto
- Department of Neurological Surgery, Okayama University, Okayama, Japan
| | - Yoshihiro Otani
- Department of Neurological Surgery, Okayama University, Okayama, Japan
| | - Ryoji Imoto
- Department of Neurological Surgery, Okayama University, Okayama, Japan
| | - Mariam Aboian
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shinichiro Koizumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kazuhiko Kurozumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Toru Kawakatsu
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kimberley Alexander
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Laveniya Satgunaseelan
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Aaron M Rulseh
- Department of Radiology, Na Homolce Hospital, Prague, Czechia
| | - Stephen J Bagley
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michel Bilello
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zev A Binder
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Steven Brem
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Arati S Desai
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert A Lustig
- Department of Radiation-Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eileen Maloney
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Timothy Prior
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nduka Amankulor
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - MacLean P Nasrallah
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Donald M O’Rourke
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Suyash Mohan
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christos Davatzikos
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Samah TAWIL, Samar MERHI. Investigating the Key Trends in Applying Artificial Intelligence to Health Technologies: A Scoping Review. PLoS One 2025; 20:e0322197. [PMID: 40372995 PMCID: PMC12080793 DOI: 10.1371/journal.pone.0322197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/18/2025] [Indexed: 05/17/2025] Open
Abstract
BACKGROUND The use of Artificial Intelligence (AI) is exponentially rising in the healthcare sector. This change influences various domains of early identification, diagnosis, and treatment of diseases. PURPOSE This study examines the integration of AI in healthcare, focusing on its transformative potential in diagnostics and treatment, and the challenges and methodologies. shaping its future development. METHODS The review included 68 academic studies retracted from different databases (WOS, Scopus and Pubmed) from January 2020 and April 2024. After careful review and data analysis, AI methodologies, benefits and challenges, were summarized. RESULTS The number of studies showed a steady rise from 2020 to 2023. Most of them were the results of a collaborative work with international universities (92.1%). The majority (66.7%) were published in top-tier (Q1) journals and 40% were cited 2-10 times. The results have shown that AI tools such as deep learning methods and machine learning continue to significantly improve accuracy and timely execution of medical processes. Benefits were discussed from both the organizational and the patient perspective in the categories of diagnosis, treatment, consultation and health monitoring of diseases. However, some challenges may exist, despite these benefits, and are related to data integration, errors related to data processing and decision making, and patient safety. CONCLUSION The article examines the present status of AI in medical applications and explores its potential future applications. The findings of this review are useful for healthcare professionals to acquire deeper knowledge on the use of medical AI from design to implementation stage. However, a thorough assessment is essential to gather more insights into whether AI benefits outweigh its risks. Additionally, ethical and privacy issues need careful consideration.
Collapse
Affiliation(s)
- TAWIL Samah
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
- Institut National de Santé Publique d’Épidémiologie Clinique et de Toxicologie-Liban (INSPECT-LB), Beirut, Lebanon
| | - MERHI Samar
- Faculty of Nursing and Health Sciences, Notre Dame University-Louaize (NDU), Zouk Mosbeh, Lebanon
| |
Collapse
|
15
|
Volovăț CC, Buzea CG, Boboc DI, Ostafe MR, Agop M, Ochiuz L, Burlea ȘL, Rusu DI, Bujor L, Iancu DT, Volovăț SR. Hybrid Deep Learning for Survival Prediction in Brain Metastases Using Multimodal MRI and Clinical Data. Diagnostics (Basel) 2025; 15:1242. [PMID: 40428235 PMCID: PMC12109748 DOI: 10.3390/diagnostics15101242] [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: 04/26/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Survival prediction in patients with brain metastases remains a major clinical challenge, where timely and individualized prognostic estimates are critical for guiding treatment strategies and patient counseling. Methods: We propose a novel hybrid deep learning framework that integrates volumetric MRI-derived imaging biomarkers with structured clinical and demographic data to predict overall survival time. Our dataset includes 148 patients from three institutions, featuring expert-annotated segmentations of enhancing tumors, necrosis, and peritumoral edema. Two convolutional neural network backbones-ResNet-50 and EfficientNet-B0-were fused with fully connected layers processing tabular data. Models were trained using mean squared error loss and evaluated through stratified cross-validation and an independent held-out test set. Results: The hybrid model based on EfficientNet-B0 achieved state-of-the-art performance, attaining an R2 score of 0.970 and a mean absolute error of 3.05 days on the test set. Permutation feature importance highlighted edema-to-tumor ratio and enhancing tumor volume as the most informative predictors. Grad-CAM visualizations confirmed the model's attention to anatomically and clinically relevant regions. Performance consistency across validation folds confirmed the framework's robustness and generalizability. Conclusions: This study demonstrates that multimodal deep learning can deliver accurate, explainable, and clinically actionable survival predictions in brain metastases. The proposed framework offers a promising foundation for integration into real-world oncology workflows to support personalized prognosis and informed therapeutic decision-making.
Collapse
Affiliation(s)
| | - Călin Gheorghe Buzea
- “Prof. Dr. Nicolae Oblu” Clinical Emergency Hospital Iași, 700309 Iași, Romania;
- National Institute of Research and Development for Technical Physics, IFT Iași, 700050 Iași, Romania
| | - Diana-Ioana Boboc
- Medical Oncology-Radiotherapy Department, “Grigore T. Popa” University of Medicine and Pharmacy Iași, 700115 Iași, Romania; (D.-I.B.); (M.-R.O.); (S.R.V.)
| | - Mădălina-Raluca Ostafe
- Medical Oncology-Radiotherapy Department, “Grigore T. Popa” University of Medicine and Pharmacy Iași, 700115 Iași, Romania; (D.-I.B.); (M.-R.O.); (S.R.V.)
| | - Maricel Agop
- Physics Department, “Gheorghe Asachi” Technical University Iași, 700050 Iași, Romania;
| | - Lăcrămioara Ochiuz
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iași, 700115 Iași, Romania; (L.O.); (Ș.L.B.)
| | - Ștefan Lucian Burlea
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iași, 700115 Iași, Romania; (L.O.); (Ș.L.B.)
| | - Dragoș Ioan Rusu
- Department of Environmental Engineering, Mechanical Engineering, Faculty of Engineering, “V. Alecsandri” University of Bacău, 600115 Bacău, Romania;
| | - Laurențiu Bujor
- Amethyst Radiotherapy, Drumul Odăii nr. 42, 719241 Otopeni, Romania;
| | - Dragoș Teodor Iancu
- Medical Oncology-Radiotherapy Department, “Grigore T. Popa” University of Medicine and Pharmacy Iași, 700115 Iași, Romania; (D.-I.B.); (M.-R.O.); (S.R.V.)
| | - Simona Ruxandra Volovăț
- Medical Oncology-Radiotherapy Department, “Grigore T. Popa” University of Medicine and Pharmacy Iași, 700115 Iași, Romania; (D.-I.B.); (M.-R.O.); (S.R.V.)
| |
Collapse
|
16
|
Sun Z, Gao J, Yu W, Yuan X, Du P, Chen P, Wang Y. Personalized prediction of breast cancer candidates for Anti-HER2 therapy using 18F-FDG PET/CT parameters and machine learning: a dual-center study. Front Oncol 2025; 15:1590769. [PMID: 40438696 PMCID: PMC12116446 DOI: 10.3389/fonc.2025.1590769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Accepted: 04/23/2025] [Indexed: 06/01/2025] Open
Abstract
Background Accurately evaluating human epidermal growth factor receptor (HER2) expression status in breast cancer enables clinicians to develop individualized treatment plans and improve patient prognosis. The purpose of this study was to assess the performance of a machine learning (ML) model that was developed using 18F-FDG PET/CT parameters and clinicopathological features in distinguishing different levels of HER2 expression in breast cancer. Methods This retrospective study enrolled breast cancer patients who underwent 18F-FDG PET/CT scans prior to treatment at Lianyungang First People's Hospital (centre 1, n=157) and the Third Affiliated Hospital of Soochow University (centre 2, n=84). Two classification tasks were analysed: distinguishing HER2-zero expression from HER2-low/positive expression (Task 1) and distinguishing HER2-low expression from HER2-positive expression (Task 2). For each task, patients from Centre 1 were randomly divided into training and internal test sets at a 7:3 ratio, whereas patients from Centre 2 served as an external test set. The prediction models included logistic regression (LR), support vector machine (SVM), extreme gradient boosting (XGBoost) and multilayer perceptron (MLP), and SHAP analysis provided model interpretability. Model performance was evaluated via the area under the receiver operating characteristic curve (AUC), accuracy, sensitivity, specificity, positive predictive value (PPV) and negative predictive value (NPV). Results XGBoost models exhibited the best predictive performance in both tasks. For Task 1, recursive feature elimination (RFE) was used to select 8 features, excluding pathological features, and the XGBoost model achieved AUCs of 0.888, 0.844 and 0.759 for the training, internal and external testing sets, respectively. The top three features according to the SHAP values were the tumour minimum diameter, mean standardized uptake value (SUVmean) and CTmean. For Task 2, 9 features were selected, including progesterone receptor (PR) status as a pathological feature. The XGBoost model achieved AUCs of 0.920, 0.814 and 0.693 for the training, internal and external testing sets, respectively. The top three features according to the SHAP values were the PR status, maximum tumour diameter and metabolic tumour volume (MTV). Conclusions ML models that incorporate 18F-FDG PET/CT parameters and clinicopathological features can aid in the prediction of different HER2 expression statuses in breast cancer.
Collapse
Affiliation(s)
- Zhenguo Sun
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jianxiong Gao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Wenji Yu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiaoshuai Yuan
- Department of Nuclear Medicine, The First People’s Hospital of Lianyungang/The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Peng Du
- Department of Nuclear Medicine, The First People’s Hospital of Lianyungang/The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Peng Chen
- Department of Nuclear Medicine, The First People’s Hospital of Lianyungang/The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Yuetao Wang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
17
|
Yang H, Zhang Y, Li F, Liu W, Zeng H, Yuan H, Ye Z, Huang Z, Yuan Y, Xiang Y, Wu K, Liu H. CT-based AI framework leveraging multi-scale features for predicting pathological grade and Ki67 index in clear cell renal cell carcinoma: a multicenter study. Insights Imaging 2025; 16:102. [PMID: 40369234 PMCID: PMC12078187 DOI: 10.1186/s13244-025-01980-0] [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: 07/22/2024] [Accepted: 03/30/2025] [Indexed: 05/16/2025] Open
Abstract
PURPOSE To explore whether a CT-based AI framework, leveraging multi-scale features, can offer a non-invasive approach to accurately predict pathological grade and Ki67 index in clear cell renal cell carcinoma (ccRCC). METHODS In this multicenter retrospective study, a total of 1073 pathologically confirmed ccRCC patients from seven cohorts were split into internal cohorts (training and validation sets) and an external test set. The AI framework comprised an image processor, a 3D-kidney and tumor segmentation model by 3D-UNet, a multi-scale features extractor built upon unsupervised learning, and a multi-task classifier utilizing XGBoost. A quantitative model interpretation technique, known as SHapley Additive exPlanations (SHAP), was employed to explore the contribution of multi-scale features. RESULTS The 3D-UNet model showed excellent performance in segmenting both the kidney and tumor regions, with Dice coefficients exceeding 0.92. The proposed multi-scale features model exhibited strong predictive capability for pathological grading and Ki67 index, with AUROC values of 0.84 and 0.87, respectively, in the internal validation set, and 0.82 and 0.82, respectively, in the external test set. The SHAP results demonstrated that features from radiomics, the 3D Auto-Encoder, and dimensionality reduction all made significant contributions to both prediction tasks. CONCLUSIONS The proposed AI framework, leveraging multi-scale features, accurately predicts the pathological grade and Ki67 index of ccRCC. CRITICAL RELEVANCE STATEMENT The CT-based AI framework leveraging multi-scale features offers a promising avenue for accurately predicting the pathological grade and Ki67 index of ccRCC preoperatively, indicating a direction for non-invasive assessment. KEY POINTS Non-invasively determining pathological grade and Ki67 index in ccRCC could guide treatment decisions. The AI framework integrates segmentation, classification, and model interpretation, enabling fully automated analysis. The AI framework enables non-invasive preoperative detection of high-risk tumors, assisting clinical decision-making.
Collapse
Affiliation(s)
- Huancheng Yang
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Radiology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
| | - Yueyue Zhang
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fan Li
- Department of Radiology, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, China
| | - Weihao Liu
- Shantou University Medical College, Shantou University, Shantou, China
| | - Haoyang Zeng
- Shantou University Medical College, Shantou University, Shantou, China
| | - Haoyuan Yuan
- Shantou University Medical College, Shantou University, Shantou, China
| | - Zixi Ye
- Shantou University Medical College, Shantou University, Shantou, China
| | - Zexin Huang
- Department of Radiology, Shenzhen Luohu District Traditional Chinese Medicine Hospital (Luohu Hospital Group), Shenzhen, China
| | - Yangguang Yuan
- Department of Radiology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
| | - Ye Xiang
- Department of Radiology, Leshan Hospital, Chengdu University of Traditional Chinese Medicine, Leshan, China.
| | - Kai Wu
- Department of Radiology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China.
| | - Hanlin Liu
- Department of Radiology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China.
| |
Collapse
|
18
|
Xu J, Miao JG, Wang CX, Zhu YP, Liu K, Qin SY, Chen HS, Lang N. CT-based quantification of intratumoral heterogeneity for predicting distant metastasis in retroperitoneal sarcoma. Insights Imaging 2025; 16:99. [PMID: 40346399 PMCID: PMC12064543 DOI: 10.1186/s13244-025-01977-9] [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: 02/05/2025] [Accepted: 04/23/2025] [Indexed: 05/11/2025] Open
Abstract
OBJECTIVES Retroperitoneal sarcoma (RPS) is highly heterogeneous, leading to different risks of distant metastasis (DM) among patients with the same clinical stage. This study aims to develop a quantitative method for assessing intratumoral heterogeneity (ITH) using preoperative contrast-enhanced CT (CECT) scans and evaluate its ability to predict DM risk. METHODS We conducted a retrospective analysis of 274 PRS patients who underwent complete surgical resection and were monitored for ≥ 36 months at two centers. Conventional radiomics (C-radiomics), ITH radiomics, and deep-learning (DL) features were extracted from the preoperative CECT scans and developed single-modality models. Clinical indicators and high-throughput CECT features were integrated to develop a combined model for predicting DM. The performance of the models was evaluated by measuring the receiver operating characteristic curve and Harrell's concordance index (C-index). Distant metastasis-free survival (DMFS) was also predicted to further assess survival benefits. RESULTS The ITH model demonstrated satisfactory predictive capability for DM in internal and external validation cohorts (AUC: 0.735, 0.765; C-index: 0.691, 0.729). The combined model that combined clinicoradiological variables, ITH-score, and DL-score achieved the best predictive performance in internal and external validation cohorts (AUC: 0.864, 0.801; C-index: 0.770, 0.752), successfully stratified patients into high- and low-risk groups for DM (p < 0.05). CONCLUSIONS The combined model demonstrated promising potential for accurately predicting the DM risk and stratifying the DMFS risk in RPS patients undergoing complete surgical resection, providing a valuable tool for guiding treatment decisions and follow-up strategies. CRITICAL RELEVANCE STATEMENT The intratumoral heterogeneity analysis facilitates the identification of high-risk retroperitoneal sarcoma patients prone to distant metastasis and poor prognoses, enabling the selection of candidates for more aggressive surgical and post-surgical interventions. KEY POINTS Preoperative identification of retroperitoneal sarcoma (RPS) with a high potential for distant metastasis (DM) is crucial for targeted interventional strategies. Quantitative assessment of intratumoral heterogeneity achieved reasonable performance for predicting DM. The integrated model combining clinicoradiological variables, ITH radiomics, and deep-learning features effectively predicted distant metastasis-free survival.
Collapse
Affiliation(s)
- Jun Xu
- Department of Radiology, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing, China
| | - Jian-Guo Miao
- The College of Computer Science & Technology, Qingdao University, No. 308, Ning Xia Road, Shinan District, Qingdao, Shandong, China
| | - Chen-Xi Wang
- Department of Radiology, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing, China
| | - Yu-Peng Zhu
- Department of Radiology, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing, China
| | - Ke Liu
- Department of Radiology, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing, China
| | - Si-Yuan Qin
- Department of Radiology, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing, China
| | - Hai-Song Chen
- Department of Radiology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, Shandong, China.
| | - Ning Lang
- Department of Radiology, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing, China.
| |
Collapse
|
19
|
Yan Y, Liu Y, Wang Y, Jiang T, Xie J, Zhou Y, Liu X, Yan M, Zheng Q, Xu H, Chen J, Sui L, Chen C, Ru R, Wang K, Zhao A, Li S, Zhu Y, Zhang Y, Wang VY, Xu D. Hierarchical diagnosis of breast phyllodes tumors enabled by deep learning of ultrasound images: a retrospective multi-center study. Cancer Imaging 2025; 25:61. [PMID: 40340752 PMCID: PMC12063467 DOI: 10.1186/s40644-025-00879-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
OBJECTIVE Phyllodes tumors (PTs) are rare breast tumors with high recurrence rates, current methods relying on post-resection pathology often delay detection and require further surgery. We propose a deep-learning-based Phyllodes Tumors Hierarchical Diagnosis Model (PTs-HDM) for preoperative identification and grading. METHODS Ultrasound images from five hospitals were retrospectively collected, with all patients having undergone surgical pathological confirmation of either PTs or fibroadenomas (FAs). PTs-HDM follows a two-stage classification: first distinguishing PTs from FAs, then grading PTs into benign or borderline/malignant. Model performance metrics including AUC and accuracy were quantitatively evaluated. A comparative analysis was conducted between the algorithm's diagnostic capabilities and those of radiologists with varying clinical experience within an external validation cohort. Through the provision of PTs-HDM's automated classification outputs and associated thermal activation mapping guidance, we systematically assessed the enhancement in radiologists' diagnostic concordance and classification accuracy. RESULTS A total of 712 patients were included. On the external test set, PTs-HDM achieved an AUC of 0.883, accuracy of 87.3% for PT vs. FA classification. Subgroup analysis showed high accuracy for tumors < 2 cm (90.9%). In hierarchical classification, the model obtained an AUC of 0.856 and accuracy of 80.9%. Radiologists' performance improved with PTs-HDM assistance, with binary classification accuracy increasing from 82.7%, 67.7%, and 64.2-87.6%, 76.6%, and 82.1% for senior, attending, and resident radiologists, respectively. Their hierarchical classification AUCs improved from 0.566 to 0.827 to 0.725-0.837. PTs-HDM also enhanced inter-radiologist consistency, increasing Kappa values from - 0.05 to 0.41 to 0.12 to 0.65, and the intraclass correlation coefficient from 0.19 to 0.45. CONCLUSION PTs-HDM shows strong diagnostic performance, especially for small lesions, and improves radiologists' accuracy across all experience levels, bridging diagnostic gaps and providing reliable support for PTs' hierarchical diagnosis.
Collapse
Affiliation(s)
- Yuqi Yan
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, No.1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China
- Wenling Institute of Big Data and Artificial Intelligence Institute in Medicine, No.18, Civic Avenue, Wenling, Taizhou, Zhejiang, 317502, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Branch of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou, Zhejiang, China
- Center of Intelligent Diagnosis and Therapy (Taizhou), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Taizhou, Zhejiang, China
- Interventional Medicine and Engineering Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Yuanzhen Liu
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, No.1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China
| | - Yao Wang
- Department of Ultrasound, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Tian Jiang
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, No.1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China
- Interventional Medicine and Engineering Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Jiayu Xie
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Yahan Zhou
- Wenling Institute of Big Data and Artificial Intelligence Institute in Medicine, No.18, Civic Avenue, Wenling, Taizhou, Zhejiang, 317502, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Branch of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou, Zhejiang, China
- Center of Intelligent Diagnosis and Therapy (Taizhou), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Taizhou, Zhejiang, China
| | - Xin Liu
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, No.1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China
| | - Meiying Yan
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, No.1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China
| | - Qiuqing Zheng
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, No.1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China
| | - Haifei Xu
- Department of Ultrasound, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Jinxiao Chen
- Department of Ultrasound, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Lin Sui
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, No.1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China
- Wenling Institute of Big Data and Artificial Intelligence Institute in Medicine, No.18, Civic Avenue, Wenling, Taizhou, Zhejiang, 317502, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Branch of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou, Zhejiang, China
- Center of Intelligent Diagnosis and Therapy (Taizhou), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Taizhou, Zhejiang, China
- Interventional Medicine and Engineering Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Chen Chen
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, No.1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China
| | - RongRong Ru
- Department of Ultrasound, Zhejiang Xiaoshan Hospital, Hangzhou, Zhejiang, China
| | - Kai Wang
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Anli Zhao
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Shiyan Li
- Department of Ultrasound in Medicine, Affiliated Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying Zhu
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No.197, Ruijin 2nd Road, Huangpu District, Shanghai, Zhejiang, 200025, China.
| | - Yang Zhang
- Wenling Institute of Big Data and Artificial Intelligence Institute in Medicine, No.18, Civic Avenue, Wenling, Taizhou, Zhejiang, 317502, China.
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Branch of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou, Zhejiang, China.
- Center of Intelligent Diagnosis and Therapy (Taizhou), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Taizhou, Zhejiang, China.
| | - Vicky Yang Wang
- Wenling Institute of Big Data and Artificial Intelligence Institute in Medicine, No.18, Civic Avenue, Wenling, Taizhou, Zhejiang, 317502, China.
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Branch of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou, Zhejiang, China.
- Center of Intelligent Diagnosis and Therapy (Taizhou), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Taizhou, Zhejiang, China.
| | - Dong Xu
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, No.1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang, 310022, China.
- Wenling Institute of Big Data and Artificial Intelligence Institute in Medicine, No.18, Civic Avenue, Wenling, Taizhou, Zhejiang, 317502, China.
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Branch of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou, Zhejiang, China.
- Center of Intelligent Diagnosis and Therapy (Taizhou), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Taizhou, Zhejiang, China.
- Interventional Medicine and Engineering Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| |
Collapse
|
20
|
Chen D, He E, Pace K, Chekay M, Raman S. Concordance with SPIRIT-AI guidelines in reporting of randomized controlled trial protocols investigating artificial intelligence in oncology: a systematic review. Oncologist 2025; 30:oyaf112. [PMID: 40421957 PMCID: PMC12107541 DOI: 10.1093/oncolo/oyaf112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/24/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Artificial intelligence (AI) is a promising tool used in oncology that may be able to facilitate diagnosis, treatment planning, and patient management. Transparency and completeness of protocols of randomized controlled trials (RCT) involving AI interventions is necessary to ensure reproducibility of AI tools across diverse clinical settings. The SPIRIT 2013 and SPIRIT-AI 2020 guidelines were developed as evidence-based recommendations for complete reporting of trial protocols. However, the concordance of AI RCT protocols in oncology to SPIRIT reporting guidelines remains unknown. This systematic review evaluates the concordance of protocols of RCTs evaluating AI interventions in oncology to the SPIRIT 2013 and SPIRIT-AI 2020 reporting guidelines. METHODS A systematic search of Ovid Medline and Embase was conducted on October 22, 2024 for primary, peer-reviewed RCT protocols involving AI interventions in oncology. Eligible studies were screened in duplicate and data extraction assessed concordance to SPIRIT 2013 and SPIRIT-AI 2020 guideline items. Item-specific concordance was measured as the proportion of studies that reported the item. Average concordance was measured as the median proportion of items reported for each study. RESULTS Twelve RCT protocols met the inclusion criteria. The median concordance to SPIRIT 2013 guidelines was 81.92% (IQR 74.88-88.95) and the median concordance to SPIRIT-AI 2020 guidelines was 78.21% (IQR 67.21-89.20). For SPIRIT 2013 guidelines, high concordance was observed for items related to study objectives and ethics, but gaps were identified in reporting blinding procedures, participant retention, and post-trial care. For SPIRIT-AI 2020 guidelines, there remained gaps based on data quality management, performance error analysis, and accessibility of AI intervention code. CONCLUSION While concordance to reporting guidelines in oncology AI RCT protocols was moderately high, critical gaps in protocol reporting persist that may hinder reproducibility and clinical implementation. Future efforts should focus on increasing awareness and reinforcement to enhance reporting quality necessary to foster the responsible integration of AI into oncology practice.
Collapse
Affiliation(s)
- David Chen
- Princess Margaret Hospital Cancer Centre, Radiation Medicine Program, Toronto, Ontario, Canada M5G 2C4
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada M5S 3K3
| | - Emily He
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada M5S 3K3
| | - Keiran Pace
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada M5S 3K3
| | - Matthew Chekay
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada M5S 3K3
| | - Srinivas Raman
- Princess Margaret Hospital Cancer Centre, Radiation Medicine Program, Toronto, Ontario, Canada M5G 2C4
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada M5T 1P5
- Department of Radiation Oncology, BC Cancer Vancouver, Vancouver, BC, Canada V5Z 1M9
| |
Collapse
|
21
|
Chadha S, Mukherjee S, Sanyal S. Advancements and implications of artificial intelligence for early detection, diagnosis and tailored treatment of cancer. Semin Oncol 2025; 52:152349. [PMID: 40345002 DOI: 10.1016/j.seminoncol.2025.152349] [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: 01/06/2025] [Revised: 03/20/2025] [Accepted: 04/04/2025] [Indexed: 05/11/2025]
Abstract
The complexity and heterogeneity of cancer makes early detection and effective treatment crucial to enhance patient survival and quality of life. The intrinsic creative ability of artificial intelligence (AI) offers improvements in patient screening, diagnosis, and individualized care. Advanced technologies, like computer vision, machine learning, deep learning, and natural language processing, can analyze large datasets and identify patterns that permit early cancer detection, diagnosis, management and incorporation of conclusive treatment plans, ensuring improved quality of life for patients by personalizing care and minimizing unnecessary interventions. Genomics, transcriptomics and proteomics data can be combined with AI algorithms to unveil an extensive overview of cancer biology, assisting in its detailed understanding and will help in identifying new drug targets and developing effective therapies. This can also help to identify personalized molecular signatures which can facilitate tailored interventions addressing the unique aspects of each patient. AI-driven transcriptomics, proteomics, and genomes represents a revolutionary strategy to improve patient outcome by offering precise diagnosis and tailored therapy. The inclusion of AI in oncology may boost efficiency, reduce errors, and save costs, but it cannot take the role of medical professionals. While clinicians and doctors have the final say in all matters, it might serve as their faithful assistant.
Collapse
Affiliation(s)
- Sonia Chadha
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, Uttar Pradesh, India.
| | - Sayali Mukherjee
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, Uttar Pradesh, India
| | - Somali Sanyal
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, Uttar Pradesh, India
| |
Collapse
|
22
|
Sguanci M, Palomares SM, Cangelosi G, Petrelli F, Sandri E, Ferrara G, Mancin S. Artificial Intelligence in the Management of Malnutrition in Cancer Patients: A Systematic Review. Adv Nutr 2025:100438. [PMID: 40334987 DOI: 10.1016/j.advnut.2025.100438] [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: 02/11/2025] [Revised: 04/23/2025] [Accepted: 04/28/2025] [Indexed: 05/09/2025] Open
Abstract
Malnutrition is a critical complication among cancer patients, affecting ≤80% of individuals depending on cancer type, stage, and treatment. Artificial intelligence (AI) has emerged as a promising tool in healthcare, with potential applications in nutritional management to improve early detection, risk stratification, and personalized interventions. This systematic review evaluated the role of AI in identifying and managing malnutrition in cancer patients, focusing on its effectiveness in nutritional status assessment, prediction, clinical outcomes, and body composition monitoring. A systematic search was conducted across PubMed, Cochrane Library, Cumulative Index to Nursing and Allied Health Literature, and Excerpta Medica Database from June to July 2024, following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Quantitative primary studies investigating AI-based interventions for malnutrition detection, body composition analysis, and nutritional optimization in oncology were included. Study quality was assessed using the Joanna Briggs Institute Critical Appraisal Tools, and evidence certainty was evaluated with the Oxford Centre for Evidence-Based Medicine framework. Eleven studies (n = 52,228 patients) met the inclusion criteria and were categorized into 3 overarching domains: nutritional status assessment and prediction, clinical and functional outcomes, and body composition and cachexia monitoring. AI-based models demonstrated high predictive accuracy in malnutrition detection (area under the curve >0.80). Machine learning algorithms, including decision trees, random forests, and support vector machines, outperformed conventional screening tools. Deep learning models applied to medical imaging achieved high segmentation accuracy (Dice similarity coefficient: 0.92-0.94), enabling early cachexia detection. AI-driven virtual dietitian systems improved dietary adherence (84%) and reduced unplanned hospitalizations. AI-enhanced workflows streamlined dietitian referrals, reducing referral times by 2.4 d. AI demonstrates significant potential in optimizing malnutrition screening, body composition monitoring, and personalized nutritional interventions for cancer patients. Its integration into oncology nutrition care could enhance patient outcomes and optimize healthcare resource allocation. Further research is necessary to standardize AI models and ensure clinical applicability. This systematic review followed a protocol registered prospectively on Open Science Framework (https://doi.org/10.17605/OSF.IO/A259M).
Collapse
Affiliation(s)
| | - Sara Morales Palomares
- Department of Pharmacy, Health and Nutritional Sciences (DFSSN), University of Calabria, Rende, Italy
| | - Giovanni Cangelosi
- School of Pharmacy, Polo Medicina Sperimentale e Sanità Pubblica "Stefania Scuri," Camerino, Italy
| | - Fabio Petrelli
- School of Pharmacy, Polo Medicina Sperimentale e Sanità Pubblica "Stefania Scuri," Camerino, Italy
| | - Elena Sandri
- Faculty of Medicine and Health Sciences, Catholic University of Valencia San Vicente Mártir, c/Quevedo, Valencia, Spain.
| | - Gaetano Ferrara
- Nephrology and Dialysis Unit, Ramazzini Hospital, Carpi, Italy
| | - Stefano Mancin
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089, Rozzano, Milan, Italy
| |
Collapse
|
23
|
Xie Y, Wang F, Wei J, Shen Z, Song X, Wang Y, Chen H, Tao L, Zheng J, Lin L, Niu Z, Guan X, Zhou T, Xu Z, Liu Y, Du D, Pan H, Li S, Ji W, Zhou W, Yang Y, Tian J, Xu J, Hu H, Liang X. Noninvasive prognostic classification of ITH in HCC with multi-omics insights and therapeutic implications. SCIENCE ADVANCES 2025; 11:eads8323. [PMID: 40315307 PMCID: PMC12047409 DOI: 10.1126/sciadv.ads8323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 03/31/2025] [Indexed: 05/04/2025]
Abstract
Intratumoral heterogeneity (ITH) is a critical factor associated with treatment failure and disease relapse in hepatocellular carcinoma (HCC). However, decoding ITH in a noninvasive and comprehensive manner remains a notable challenge. In this study involving 851 patients from five centers, we developed a noninvasive prognostic classification for ITH using radiomics based on multisequence MRI, termed radiomics ITH (RITH) phenotypes. The RITH phenotypes highly correlated with prognosis and pathological ITH. In addition, through an integrated multi-omics analysis, we uncovered the molecular mechanisms underlying RITH, notably enhancing its biological interpretability. Specifically, high-RITH tumors demonstrated an enrichment of cancer-associated fibroblasts and activation of extracellular matrix remodeling. Our approach facilitates the noninvasive refined classification of ITH using radiomics and multi-omics, paving the way for tailored treatment strategies in HCC. Extracellular matrix-receptor interaction could be a potential therapeutic target in patients with high-RITH tumors. Given the routine use of radiologic imaging in oncology, our methodology ignites versatile framework for broader application to other solid tumors.
Collapse
Affiliation(s)
- Yangyang Xie
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Fang Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, 430022 Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, 430022 Wuhan, China
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
| | - Jingwei Wei
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 100190 Beijing, China
- Beijing Key Laboratory of Molecular Imaging, 100190 Beijing, China
| | - Zefeng Shen
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Xue Song
- Department of Respiratory and Critical Care Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 310007 Hangzhou, China
| | - Yali Wang
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Hongjun Chen
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Liye Tao
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Junhao Zheng
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Lanfen Lin
- The College of Computer Science and Technology, Zhejiang University, 310027 Hangzhou, China
| | - Ziwei Niu
- The College of Computer Science and Technology, Zhejiang University, 310027 Hangzhou, China
| | - Xiaojun Guan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009 Hangzhou, China
| | - Tianhan Zhou
- Department of General Surgery, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 310007 Hangzhou, China
| | - Zhengao Xu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Yang Liu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Danwei Du
- Department of Anorectal, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 310000 Hangzhou, China
| | - Haoyu Pan
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Shihao Li
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Wenbin Ji
- Department of Radiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 317000 Taizhou, China
| | - Wei Zhou
- Department of Radiology, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, 313000 Huzhou, China
| | - Yunjun Yang
- Department of Radiology, The First Affiliated Hospital, Wenzhou Medical University, 325006 Wenzhou, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 100190 Beijing, China
- Beijing Key Laboratory of Molecular Imaging, 100190 Beijing, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 100191 Beijing, China
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, 710126 Xi’an, China
| | - Junjie Xu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Hongjie Hu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
| | - Xiao Liang
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- School of Medicine, Shaoxing University, 312000 Shaoxing, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, 310000 Hangzhou, China
| |
Collapse
|
24
|
Chinta SV, Wang Z, Palikhe A, Zhang X, Kashif A, Smith MA, Liu J, Zhang W. AI-driven healthcare: Fairness in AI healthcare: A survey. PLOS DIGITAL HEALTH 2025; 4:e0000864. [PMID: 40392801 PMCID: PMC12091740 DOI: 10.1371/journal.pdig.0000864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Artificial intelligence (AI) is rapidly advancing in healthcare, enhancing the efficiency and effectiveness of services across various specialties, including cardiology, ophthalmology, dermatology, emergency medicine, etc. AI applications have significantly improved diagnostic accuracy, treatment personalization, and patient outcome predictions by leveraging technologies such as machine learning, neural networks, and natural language processing. However, these advancements also introduce substantial ethical and fairness challenges, particularly related to biases in data and algorithms. These biases can lead to disparities in healthcare delivery, affecting diagnostic accuracy and treatment outcomes across different demographic groups. This review paper examines the integration of AI in healthcare, highlighting critical challenges related to bias and exploring strategies for mitigation. We emphasize the necessity of diverse datasets, fairness-aware algorithms, and regulatory frameworks to ensure equitable healthcare delivery. The paper concludes with recommendations for future research, advocating for interdisciplinary approaches, transparency in AI decision-making, and the development of innovative and inclusive AI applications.
Collapse
Affiliation(s)
| | - Zichong Wang
- Florida International University, Miami, Florida, United States of America
| | - Avash Palikhe
- Florida International University, Miami, Florida, United States of America
| | - Xingyu Zhang
- University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Ayesha Kashif
- Jose Marti MAST 6-12 Academy, Hialeah, Florida, United States of America
| | | | - Jun Liu
- Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Wenbin Zhang
- Florida International University, Miami, Florida, United States of America
| |
Collapse
|
25
|
Kayaalp ME, Prill R, Sezgin EA, Cong T, Królikowska A, Hirschmann MT. DeepSeek versus ChatGPT: Multimodal artificial intelligence revolutionizing scientific discovery. From language editing to autonomous content generation-Redefining innovation in research and practice. Knee Surg Sports Traumatol Arthrosc 2025; 33:1553-1556. [PMID: 39936363 DOI: 10.1002/ksa.12628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/13/2025]
Affiliation(s)
- Mahmut Enes Kayaalp
- Department of Orthopaedics and Traumatology, University of Health Sciences, Istanbul Fatih Sultan Mehmet Training and Research Hospital, Istanbul, Turkey
| | - Robert Prill
- Department of Orthopaedics and Traumatology, University Hospital Brandenburg/Havel, Brandenburg Medical School Theodor Fontane, Brandenburg/Havel, Germany
| | - Erdem Aras Sezgin
- Department of Orthopaedics and Traumatology, Gazi University, Ankara, Turkey
| | - Ting Cong
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Aleksandra Królikowska
- Physiotherapy Research Laboratory, University Centre of Physiotherapy and Rehabilitation, Faculty of Physiotherapy, Wroclaw Medical University, Wroclaw, Poland
| | - Michael T Hirschmann
- Department of Orthopedic Surgery and Traumatology, Kantonsspital Baselland, Bruderholz, Switzerland
- Department of Clinical Research, Research Group Michael T. Hirschmann, Regenerative Medicine & Biomechanics, University of Basel, Basel, Switzerland
| |
Collapse
|
26
|
Dhali A, Kipkorir V, Maity R, Srichawla B, Biswas J, Rathna R, Bharadwaj H, Ongidi I, Chaudhry T, Morara G, Waithaka M, Rugut C, Lemashon M, Cheruiyot I, Ojuka D, Ray S, Dhali G. Artificial Intelligence-Assisted Capsule Endoscopy Versus Conventional Capsule Endoscopy for Detection of Small Bowel Lesions: A Systematic Review and Meta-Analysis. J Gastroenterol Hepatol 2025; 40:1105-1118. [PMID: 40083189 PMCID: PMC12062924 DOI: 10.1111/jgh.16931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/04/2025] [Accepted: 03/01/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Capsule endoscopy (CE) is a valuable tool used in the diagnosis of small intestinal lesions. The study aims to systematically review the literature and provide a meta-analysis of the diagnostic accuracy, specificity, sensitivity, and negative and positive predictive values of AI-assisted CE in the diagnosis of small bowel lesions in comparison to CE. METHODS Literature searches were performed through PubMed, SCOPUS, and EMBASE to identify studies eligible for inclusion. All publications up to 24 November 2024 were included. Original articles (including observational studies and randomized control trials), systematic reviews, meta-analyses, and case series reporting outcomes on AI-assisted CE in the diagnosis of small bowel lesions were included. The extracted data were pooled, and a meta-analysis was performed for the appropriate variables, considering the clinical and methodological heterogeneity among the included studies. Comprehensive Meta-Analysis v4.0 (Biostat Inc.) was used for the analysis of the data. RESULTS A total of 14 studies were included in the present study. The mean age of participants across the studies was 54.3 years (SD 17.7), with 55.4% men and 44.6% women. The pooled accuracy for conventional CE was 0.966 (95% CI: 0.925-0.988), whereas for AI-assisted CE, it was 0.9185 (95% CI: 0.9138-0.9233). Conventional CE exhibited a pooled sensitivity of 0.860 (95% CI: 0.786-0.934) compared with AI-assisted CE at 0.9239 (95% CI: 0.8648-0.9870). The positive predictive value for conventional CE was 0.982 (95% CI: 0.976-0.987), whereas AI-assisted CE had a PPV of 0.8928 (95% CI: 0.7554-0.999). The pooled specificity for conventional CE was 0.998 (95% CI: 0.996-0.999) compared with 0.5367 (95% CI: 0.5244-0.5492) for AI-assisted CE. Negative predictive values were higher in AI-assisted CE at 0.9425 (95% CI: 0.9389-0.9462) versus 0.760 (95% CI: 0.577-0.943) for conventional CE. CONCLUSION AI-assisted CE displays superior diagnostic accuracy, sensitivity, and positive predictive values albeit the lower pooled specificity in comparison with conventional CE. Its use would ensure accurate detection of small bowel lesions and further enhance their management.
Collapse
Affiliation(s)
- Arkadeep Dhali
- Academic Unit of GastroenterologySheffield Teaching Hospitals NHS Foundation TrustSheffieldUK
- School of Medicine and Population HealthUniversity of SheffieldSheffieldUK
| | | | - Rick Maity
- Institute of Post Graduate Medical Education and ResearchKolkataIndia
| | | | | | | | | | - Ibsen Ongidi
- Faculty of Health SciencesUniversity of NairobiNairobiKenya
| | - Talha Chaudhry
- Faculty of Health SciencesUniversity of NairobiNairobiKenya
| | - Gisore Morara
- Faculty of Health SciencesUniversity of NairobiNairobiKenya
| | | | - Clinton Rugut
- Faculty of Health SciencesUniversity of NairobiNairobiKenya
| | | | | | - Daniel Ojuka
- Faculty of Health SciencesUniversity of NairobiNairobiKenya
| | - Sukanta Ray
- Institute of Post Graduate Medical Education and ResearchKolkataIndia
| | | |
Collapse
|
27
|
Aden D, Zaheer S, Sureka N, Trisal M, Chaurasia JK, Zaheer S. Exploring immune checkpoint inhibitors: Focus on PD-1/PD-L1 axis and beyond. Pathol Res Pract 2025; 269:155864. [PMID: 40068282 DOI: 10.1016/j.prp.2025.155864] [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: 08/31/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy emerges as a promising approach, marked by recent substantial progress in elucidating how the host immune response impacts tumor development and its sensitivity to various treatments. Immune checkpoint inhibitors have revolutionized cancer therapy by unleashing the power of the immune system to recognize and eradicate tumor cells. Among these, inhibitors targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have garnered significant attention due to their remarkable clinical efficacy across various malignancies. This review delves into the mechanisms of action, clinical applications, and emerging therapeutic strategies surrounding PD-1/PD-L1 blockade. We explore the intricate interactions between PD-1/PD-L1 and other immune checkpoints, shedding light on combinatorial approaches to enhance treatment outcomes and overcome resistance mechanisms. Furthermore, we discuss the expanding landscape of immune checkpoint inhibitors beyond PD-1/PD-L1, including novel targets such as CTLA-4, LAG-3, TIM-3, and TIGIT. Through a comprehensive analysis of preclinical and clinical studies, we highlight the promise and challenges of immune checkpoint blockade in cancer immunotherapy, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India.
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Monal Trisal
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
28
|
Zhang W, Zhuang D, Wei W, Yang Y, Ma L, Du H, Jin A, He J, Li X. The 100 most-cited radiomics articles in cancer research: A bibliometric analysis. Clin Imaging 2025; 121:110442. [PMID: 40086035 DOI: 10.1016/j.clinimag.2025.110442] [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: 10/02/2024] [Revised: 02/15/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Radiomics, an advanced medical imaging analysis technique introduced by Professor Lambin in 2012, has quickly become a key area of medical research. To explore emerging trends in cancer-related radiomics, we conducted a bibliometric analysis of the 100 most-cited articles (T100) in this field. Data were collected from the Web of Science Core Collection on October 7, 2023, and the articles were ranked by citation count. We extracted data such as authors, journals, citation counts, and publication years and analyzed it using Microsoft Excel 2019 and R 4.4.2. CiteSpace was used to create co-occurrence and citation burst maps to show the relationships between authors, countries, institutions, and keywords. The analysis revealed that the T100 came from 81 countries, with the U.S. contributing the most (56 articles). Harvard University was the leading institution, and the journal Radiology had the highest citation count. Aerts Hugo JWL was the most influential author. The study highlights that "lung cancer" and "artificial intelligence" are emerging as major research hotspots, shaping the future of cancer radiomics.
Collapse
Affiliation(s)
- Wenhao Zhang
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China; Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China; Department of Clinical Medical, First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Dongmei Zhuang
- Suzhou Hospital of Anhui Medical University, Suzhou, Anhui, China
| | - Wenzhuo Wei
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - Yuchen Yang
- Department of Clinical Medical, First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Lijun Ma
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - He Du
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - Anran Jin
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - Jingyi He
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - Xiaoming Li
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China; Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China.
| |
Collapse
|
29
|
Missaoui R, Hechkel W, Saadaoui W, Helali A, Leo M. Advanced Deep Learning and Machine Learning Techniques for MRI Brain Tumor Analysis: A Review. SENSORS (BASEL, SWITZERLAND) 2025; 25:2746. [PMID: 40363185 PMCID: PMC12074157 DOI: 10.3390/s25092746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/09/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025]
Abstract
A brain tumor is the result of abnormal growth of cells in the central nervous system (CNS), widely considered as a complex and diverse clinical entity that is difficult to diagnose and cure. In this study, we focus on current advances in medical imaging, particularly magnetic resonance imaging (MRI), and how machine learning (ML) and deep learning (DL) algorithms might be combined with clinical assessments to improve brain tumor diagnosis. Due to its superior contrast resolution and safety compared to other imaging methods, MRI is highlighted as the preferred imaging modality for brain tumors. The challenges related to brain tumor analysis in different processes including detection, segmentation, classification, and survival prediction are addressed along with how ML/DL approaches significantly improve these steps. We systematically analyzed 107 studies (2018-2024) employing ML, DL, and hybrid models across publicly available datasets such as BraTS, TCIA, and Figshare. In the light of recent developments in brain tumor analysis, many algorithms have been proposed to accurately obtain ontological characteristics of tumors, enhancing diagnostic precision and personalized therapeutic strategies.
Collapse
Affiliation(s)
- Rim Missaoui
- Laboratory of Micro-Optoelectronics and Nanostructures (LMON), University of Monastir, Avenue of the Environment, Monastir 5019, Tunisia; (R.M.); (W.H.); (A.H.)
- National High School of Engineering of Tunis (ENSIT), 5 Rue Taha Hussein–Montfleury, Tunis 1008, Tunisia
| | - Wided Hechkel
- Laboratory of Micro-Optoelectronics and Nanostructures (LMON), University of Monastir, Avenue of the Environment, Monastir 5019, Tunisia; (R.M.); (W.H.); (A.H.)
| | - Wajdi Saadaoui
- LRMAN Laboratory, Higher Institute of Applied Sciences and Technology of Kasserine (ISSAT), Kasserine 1200, Tunisia;
| | - Abdelhamid Helali
- Laboratory of Micro-Optoelectronics and Nanostructures (LMON), University of Monastir, Avenue of the Environment, Monastir 5019, Tunisia; (R.M.); (W.H.); (A.H.)
| | - Marco Leo
- Institute of Applied Sciences and Intelligent Systems, National Research Council of Italy, 73100 Lecce, Italy
| |
Collapse
|
30
|
Ma D, Fan C, Sano T, Kawabata K, Nishikubo H, Imanishi D, Sakuma T, Maruo K, Yamamoto Y, Matsuoka T, Yashiro M. Beyond Biomarkers: Machine Learning-Driven Multiomics for Personalized Medicine in Gastric Cancer. J Pers Med 2025; 15:166. [PMID: 40423038 DOI: 10.3390/jpm15050166] [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: 03/06/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/28/2025] Open
Abstract
Gastric cancer (GC) remains one of the leading causes of cancer-related mortality worldwide, with most cases diagnosed at advanced stages. Traditional biomarkers provide only partial insights into GC's heterogeneity. Recent advances in machine learning (ML)-driven multiomics technologies, including genomics, epigenomics, transcriptomics, proteomics, metabolomics, pathomics, and radiomics, have facilitated a deeper understanding of GC by integrating molecular and imaging data. In this review, we summarize the current landscape of ML-based multiomics integration for GC, highlighting its role in precision diagnosis, prognosis prediction, and biomarker discovery for achieving personalized medicine.
Collapse
Affiliation(s)
- Dongheng Ma
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Canfeng Fan
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Tomoya Sano
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Kyoka Kawabata
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Hinano Nishikubo
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Daiki Imanishi
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Takashi Sakuma
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Koji Maruo
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Yurie Yamamoto
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Tasuku Matsuoka
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Masakazu Yashiro
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| |
Collapse
|
31
|
Guan J, Gong X, Zeng H, Zhang W, Qin Q, Gou H, Liu X, Song B. Gastrointestinal tumor personalized immunotherapy: an integrated analysis from molecular genetics to imaging biomarkers. Therap Adv Gastroenterol 2025; 18:17562848251333527. [PMID: 40297204 PMCID: PMC12035075 DOI: 10.1177/17562848251333527] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
The immunotherapy landscape for gastrointestinal (GI) tumors is rapidly evolving. There is an urgent need for reliable biomarkers capable of predicting treatment outcomes to optimize therapeutic strategies and enhance patient prognosis. This review presents a comprehensive overview of biomarkers associated with the immunotherapy response of GI tumors, covering advances in molecular genetics, histopathological markers, and imaging. Key molecular biomarkers, such as microsatellite instability, tumor mutational burden, and programmed death-ligand 1 expression, remain critical for identifying patients likely to benefit from immune checkpoint inhibitors. The significance of tumor-infiltrating lymphocytes, notably the CD8+ T cell to regulatory T cell ratio, as a predictor of immunotherapy response is explored. In addition, advanced imaging techniques, including computed tomography (CT), magnetic resonance imaging, and positron emission tomography-CT, facilitate the noninvasive evaluation of tumor biology and therapeutic response. By bridging molecular and imaging data, this integrated strategy enhances precision in patient selection, treatment monitoring, and adaptive therapy design. Future studies should aim to validate these biomarkers in larger, multicenter cohorts and focus on clinical translation to advance precision medicine in GI oncology.
Collapse
Affiliation(s)
- Jian Guan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Xiaoling Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hanjiang Zeng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Qing Qin
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongfeng Gou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xijiao Liu
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| |
Collapse
|
32
|
Saglam S, Uludag V, Karaduman ZO, Arıcan M, Yücel MO, Dalaslan RE. Comparative evaluation of artificial intelligence models GPT-4 and GPT-3.5 in clinical decision-making in sports surgery and physiotherapy: a cross-sectional study. BMC Med Inform Decis Mak 2025; 25:163. [PMID: 40229819 PMCID: PMC11998439 DOI: 10.1186/s12911-025-02996-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/07/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND The integration of artificial intelligence (AI) in healthcare has rapidly expanded, particularly in clinical decision-making. Large language models (LLMs) such as GPT-4 and GPT-3.5 have shown potential in various medical applications, including diagnostics and treatment planning. However, their efficacy in specialized fields like sports surgery and physiotherapy remains underexplored. This study aims to compare the performance of GPT-4 and GPT-3.5 in clinical decision-making within these domains using a structured assessment approach. METHODS This cross-sectional study included 56 professionals specializing in sports surgery and physiotherapy. Participants evaluated 10 standardized clinical scenarios generated by GPT-4 and GPT-3.5 using a 5-point Likert scale. The scenarios encompassed common musculoskeletal conditions, and assessments focused on diagnostic accuracy, treatment appropriateness, surgical technique detailing, and rehabilitation plan suitability. Data were collected anonymously via Google Forms. Statistical analysis included paired t-tests for direct model comparisons, one-way ANOVA to assess performance across multiple criteria, and Cronbach's alpha to evaluate inter-rater reliability. RESULTS GPT-4 significantly outperformed GPT-3.5 across all evaluated criteria. Paired t-test results (t(55) = 10.45, p < 0.001) demonstrated that GPT-4 provided more accurate diagnoses, superior treatment plans, and more detailed surgical recommendations. ANOVA results confirmed the higher suitability of GPT-4 in treatment planning (F(1, 55) = 35.22, p < 0.001) and rehabilitation protocols (F(1, 55) = 32.10, p < 0.001). Cronbach's alpha values indicated higher internal consistency for GPT-4 (α = 0.478) compared to GPT-3.5 (α = 0.234), reflecting more reliable performance. CONCLUSIONS GPT-4 demonstrates superior performance compared to GPT-3.5 in clinical decision-making for sports surgery and physiotherapy. These findings suggest that advanced AI models can aid in diagnostic accuracy, treatment planning, and rehabilitation strategies. However, AI should function as a decision-support tool rather than a substitute for expert clinical judgment. Future studies should explore the integration of AI into real-world clinical workflows, validate findings using larger datasets, and compare additional AI models beyond the GPT series.
Collapse
Affiliation(s)
- Sönmez Saglam
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Duzce University, Duzce, Türkiye.
| | - Veysel Uludag
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Duzce University, Duzce, Türkiye
| | - Zekeriya Okan Karaduman
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Duzce University, Duzce, Türkiye
| | - Mehmet Arıcan
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Duzce University, Duzce, Türkiye
| | - Mücahid Osman Yücel
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Duzce University, Duzce, Türkiye
| | - Raşit Emin Dalaslan
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Duzce University, Duzce, Türkiye
| |
Collapse
|
33
|
Huhulea EN, Huang L, Eng S, Sumawi B, Huang A, Aifuwa E, Hirani R, Tiwari RK, Etienne M. Artificial Intelligence Advancements in Oncology: A Review of Current Trends and Future Directions. Biomedicines 2025; 13:951. [PMID: 40299653 PMCID: PMC12025054 DOI: 10.3390/biomedicines13040951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 05/01/2025] Open
Abstract
Cancer remains one of the leading causes of mortality worldwide, driving the need for innovative approaches in research and treatment. Artificial intelligence (AI) has emerged as a powerful tool in oncology, with the potential to revolutionize cancer diagnosis, treatment, and management. This paper reviews recent advancements in AI applications within cancer research, focusing on early detection through computer-aided diagnosis, personalized treatment strategies, and drug discovery. We survey AI-enhanced diagnostic applications and explore AI techniques such as deep learning, as well as the integration of AI with nanomedicine and immunotherapy for cancer care. Comparative analyses of AI-based models versus traditional diagnostic methods are presented, highlighting AI's superior potential. Additionally, we discuss the importance of integrating social determinants of health to optimize cancer care. Despite these advancements, challenges such as data quality, algorithmic biases, and clinical validation remain, limiting widespread adoption. The review concludes with a discussion of the future directions of AI in oncology, emphasizing its potential to reshape cancer care by enhancing diagnosis, personalizing treatments and targeted therapies, and ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Ellen N. Huhulea
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
| | - Lillian Huang
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
| | - Shirley Eng
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
| | - Bushra Sumawi
- Barshop Institute, The University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Audrey Huang
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
| | - Esewi Aifuwa
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
| | - Rahim Hirani
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
- Graduate School of Biomedical Sciences, New York Medical College, Valhalla, NY 10595, USA
| | - Raj K. Tiwari
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
- Graduate School of Biomedical Sciences, New York Medical College, Valhalla, NY 10595, USA
| | - Mill Etienne
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
- Department of Neurology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
34
|
Liu Z, Wu Y, Xu H, Wang M, Weng S, Pei D, Chen S, Wang W, Yan J, Cui L, Duan J, Zhao Y, Wang Z, Ma Z, Li R, Duan W, Qiu Y, Su D, Li S, Liu H, Li W, Ma C, Yu M, Yu Y, Chen T, Fu J, Zhen Y, Yu B, Ji Y, Zheng H, Liang D, Liu X, Yan D, Han X, Wang F, Li ZC, Zhang Z. Multimodal fusion of radio-pathology and proteogenomics identify integrated glioma subtypes with prognostic and therapeutic opportunities. Nat Commun 2025; 16:3510. [PMID: 40222975 PMCID: PMC11994800 DOI: 10.1038/s41467-025-58675-9] [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: 08/29/2024] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
Integrating multimodal data can uncover causal features hidden in single-modality analyses, offering a comprehensive understanding of disease complexity. This study introduces a multimodal fusion subtyping (MOFS) framework that integrates radiological, pathological, genomic, transcriptomic, and proteomic data from 122 patients with IDH-wildtype adult glioma, identifying three subtypes: MOFS1 (proneural) with favorable prognosis, elevated neurodevelopmental activity, and abundant neurocyte infiltration; MOFS2 (proliferative) with the worst prognosis, superior proliferative activity, and genome instability; MOFS3 (TME-rich) with intermediate prognosis, abundant immune and stromal components, and sensitive to anti-PD-1 immunotherapy. STRAP emerges as a prognostic biomarker and potential therapeutic target for MOFS2, associated with its proliferative phenotype. Stromal infiltration in MOFS3 serves as a crucial prognostic indicator, allowing for further prognostic stratification. Additionally, we develop a deep neural network (DNN) classifier based on radiological features to further enhance the clinical translatability, providing a non-invasive tool for predicting MOFS subtypes. Overall, these findings highlight the potential of multimodal fusion in improving the classification, prognostic accuracy, and precision therapy of IDH-wildtype glioma, offering an avenue for personalized management.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China
| | - Yushuai Wu
- Shanghai Academy of Artificial Intelligence for Science, Shanghai, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Minkai Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Dongling Pei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - WeiWei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Yan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Cui
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingxian Duan
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuanshen Zhao
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zilong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zeyu Ma
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ran Li
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Wenchao Duan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuning Qiu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dingyuan Su
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sen Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haoran Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenyuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Caoyuan Ma
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Miaomiao Yu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yinhui Yu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Te Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Fu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - YingWei Zhen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin Yu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuchen Ji
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hairong Zheng
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, State Key Laboratory of Biomedical Imaging Science and System, Shenzhen, China
| | - Dong Liang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, State Key Laboratory of Biomedical Imaging Science and System, Shenzhen, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongming Yan
- Department of Neurosurgery, 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, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Fubing Wang
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Zhi-Cheng Li
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, State Key Laboratory of Biomedical Imaging Science and System, Shenzhen, China.
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
35
|
Wu C, Andaloussi MA, Hormuth DA, Lima EABF, Lorenzo G, Stowers CE, Ravula S, Levac B, Dimakis AG, Tamir JI, Brock KK, Chung C, Yankeelov TE. A critical assessment of artificial intelligence in magnetic resonance imaging of cancer. NPJ IMAGING 2025; 3:15. [PMID: 40226507 PMCID: PMC11981920 DOI: 10.1038/s44303-025-00076-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 03/17/2025] [Indexed: 04/15/2025]
Abstract
Given the enormous output and pace of development of artificial intelligence (AI) methods in medical imaging, it can be challenging to identify the true success stories to determine the state-of-the-art of the field. This report seeks to provide the magnetic resonance imaging (MRI) community with an initial guide into the major areas in which the methods of AI are contributing to MRI in oncology. After a general introduction to artificial intelligence, we proceed to discuss the successes and current limitations of AI in MRI when used for image acquisition, reconstruction, registration, and segmentation, as well as its utility for assisting in diagnostic and prognostic settings. Within each section, we attempt to present a balanced summary by first presenting common techniques, state of readiness, current clinical needs, and barriers to practical deployment in the clinical setting. We conclude by presenting areas in which new advances must be realized to address questions regarding generalizability, quality assurance and control, and uncertainty quantification when applying MRI to cancer to maintain patient safety and practical utility.
Collapse
Affiliation(s)
- Chengyue Wu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
| | | | - David A. Hormuth
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX USA
| | - Ernesto A. B. F. Lima
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
- Texas Advanced Computing Center, The University of Texas at Austin, Austin, TX USA
| | - Guillermo Lorenzo
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
- Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Casey E. Stowers
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
| | - Sriram Ravula
- Chandra Family Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX USA
| | - Brett Levac
- Chandra Family Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX USA
| | - Alexandros G. Dimakis
- Chandra Family Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX USA
| | - Jonathan I. Tamir
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
- Chandra Family Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX USA
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX USA
| | - Kristy K. Brock
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Caroline Chung
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Thomas E. Yankeelov
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX USA
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX USA
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX USA
- Department of Oncology, The University of Texas at Austin, Austin, TX USA
| |
Collapse
|
36
|
Vlad AL, Popazu C, Lescai AM, Voinescu DC, Baltă AAȘ. The Role of Artificial Intelligence in the Diagnosis and Management of Rheumatoid Arthritis. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:689. [PMID: 40282981 PMCID: PMC12028963 DOI: 10.3390/medicina61040689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: Artificial intelligence has emerged as a transformative tool in healthcare, offering capabilities such as early diagnosis, personalised treatment, and real-time patient monitoring. In the context of rheumatoid arthritis, a chronic autoimmune disease that demands timely intervention, artificial intelligence shows promise in overcoming diagnostic delays and optimising disease management. This study examines the role of artificial intelligence in the diagnosis and management of rheumatoid arthritis, focusing on perceived benefits, challenges, and acceptance levels among healthcare professionals and patients. Materials and Methods: A cross-sectional study was conducted using a detailed questionnaire distributed to 205 participants, including rheumatologists, general practitioners, and rheumatoid arthritis patients from Romania. The study used descriptive statistics, chi-square tests, and logistic regression to analyse AI acceptance in rheumatology. Data visualisation and multiple imputations addressed missing values, ensuring accuracy. Statistical significance was set at p < 0.05 for hypothesis testing. Results: Respondents with prior experience in artificial intelligence perceived it as more useful for early diagnosis and personalised management of RA (p < 0.001). Familiarity with artificial intelligence concepts positively correlated with acceptance in routine rheumatology practice (ρ = 1.066, p < 0.001). The main barriers identified were high costs (36%), lack of medical staff training (37%), and concerns regarding diagnostic accuracy (21%). Although less frequently mentioned, data privacy concerns remained relevant for a subset of respondents. The study revealed that artificial intelligence could improve diagnostic accuracy and rheumatoid arthritis monitoring, being perceived as a valuable tool by professionals familiar with digital technologies. However, 42% of participants cited the lack of data standardisation across medical systems as a major barrier, underscoring the need for effective interoperability solutions. Conclusions: Artificial intelligence has the potential to revolutionise rheumatoid arthritis management through faster and more accurate diagnoses, personalised treatments, and optimised monitoring. Nevertheless, challenges such as costs, staff training, and data privacy need to be addressed to ensure efficient integration into clinical practice. Educational programmes and interdisciplinary collaboration are essential to increase artificial intelligence adoption in rheumatology.
Collapse
Affiliation(s)
- Adriana Liliana Vlad
- Faculty of Medicine and Pharmacy, “Dunărea de Jos” University of Galați, 800008 Galați, Romania; (A.L.V.); (C.P.); (D.C.V.); (A.A.Ș.B.)
- “St. Apostle Andrei” Clinical Emergency County Hospital, 800578 Galați, Romania
| | - Corina Popazu
- Faculty of Medicine and Pharmacy, “Dunărea de Jos” University of Galați, 800008 Galați, Romania; (A.L.V.); (C.P.); (D.C.V.); (A.A.Ș.B.)
- “St. Apostle Andrei” Clinical Emergency County Hospital, 800578 Galați, Romania
| | - Alina-Maria Lescai
- Faculty of Medicine and Pharmacy, “Dunărea de Jos” University of Galați, 800008 Galați, Romania; (A.L.V.); (C.P.); (D.C.V.); (A.A.Ș.B.)
- “St. Apostle Andrei” Clinical Emergency County Hospital, 800578 Galați, Romania
| | - Doina Carina Voinescu
- Faculty of Medicine and Pharmacy, “Dunărea de Jos” University of Galați, 800008 Galați, Romania; (A.L.V.); (C.P.); (D.C.V.); (A.A.Ș.B.)
- “St. Apostle Andrei” Clinical Emergency County Hospital, 800578 Galați, Romania
| | - Alexia Anastasia Ștefania Baltă
- Faculty of Medicine and Pharmacy, “Dunărea de Jos” University of Galați, 800008 Galați, Romania; (A.L.V.); (C.P.); (D.C.V.); (A.A.Ș.B.)
- “St. Apostle Andrei” Clinical Emergency County Hospital, 800578 Galați, Romania
| |
Collapse
|
37
|
Adriaenssens N, Wuyts SCM, Steurbaut S, De Sutter PJ, Vermeulen A, de Haar-Holleman A, Beckwée D, Provyn S, Vande Casteele S, Zhou J, Lanckmans K, Van Bocxlaer J, De Nys L. Synergy of Body Composition, Exercise Oncology, and Pharmacokinetics: A Narrative Review of Personalizing Paclitaxel Treatment for Breast Cancer. Cancers (Basel) 2025; 17:1271. [PMID: 40282447 PMCID: PMC12025660 DOI: 10.3390/cancers17081271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Paclitaxel is a type of small molecule chemotherapy widely used for breast cancer, but its clinical efficacy is often hindered by dose-limiting toxicities such as chemotherapy-induced peripheral neuropathy and neutropenia. Traditional dosing based on body surface area does not account for variations in body composition, which may influence paclitaxel metabolism, toxicity, and treatment outcomes. This review explores the interplay between body composition, physical activity, and paclitaxel pharmacokinetics, emphasizing the potential for personalized dosing strategies. METHODS A comprehensive narrative review was conducted by analyzing the literature on body composition, small molecule chemotherapy-related toxicities, pharmacokinetics, and exercise oncology. Studies examining the role of skeletal muscle mass, adipose tissue, and physical activity in modulating paclitaxel metabolism and side effects were included. RESULTS Evidence suggests that patients with low skeletal muscle mass are at a higher risk of paclitaxel-induced toxicities due to altered drug distribution and clearance. Sarcopenic obesity, characterized by low muscle and high-fat levels, further exacerbates these risks. Exercise, particularly resistance and aerobic training, has been shown to improve muscle mass, mitigate toxicities, and enhance chemotherapy tolerance. However, the precise mechanisms by which exercise influences paclitaxel pharmacokinetics remain underexplored. CONCLUSIONS Personalized chemotherapy dosing, considering body composition and physical activity, may optimize paclitaxel treatment outcomes. Future research should focus on integrating exercise interventions into oncology care and refining dosing models that account for interindividual differences in drug metabolism. These advancements could improve treatment efficacy while minimizing toxicities in breast cancer patients.
Collapse
Affiliation(s)
- Nele Adriaenssens
- Rehabilitation Research, Vrije Universiteit Brussel (VUB), Laarbeeklaan 121, 1090 Brussels, Belgium (J.Z.); (L.D.N.)
- Medical Oncology Department, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Stephanie C. M. Wuyts
- Pharmacy Department, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium (S.S.)
- Research Centre for Digital Medicine, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Stephane Steurbaut
- Pharmacy Department, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium (S.S.)
- Vitality Research Group, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Pieter-Jan De Sutter
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Universiteit Gent, Ottergemsesteenweg 460, 9000 Gent, Belgium (S.V.C.)
| | - An Vermeulen
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Universiteit Gent, Ottergemsesteenweg 460, 9000 Gent, Belgium (S.V.C.)
| | - Amy de Haar-Holleman
- Medical Oncology Department, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
- Translational Oncology Research Center, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - David Beckwée
- Rehabilitation Research, Vrije Universiteit Brussel (VUB), Laarbeeklaan 121, 1090 Brussels, Belgium (J.Z.); (L.D.N.)
| | - Steven Provyn
- Human Physiology and Sports Physiotherapy, Vrije Universiteit Brussel (VUB), Pleinlaan 2, 1050 Brussels, Belgium
| | - Sofie Vande Casteele
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Universiteit Gent, Ottergemsesteenweg 460, 9000 Gent, Belgium (S.V.C.)
| | - Jinyu Zhou
- Rehabilitation Research, Vrije Universiteit Brussel (VUB), Laarbeeklaan 121, 1090 Brussels, Belgium (J.Z.); (L.D.N.)
| | - Katrien Lanckmans
- Clinical Biology Department, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium;
| | - Jan Van Bocxlaer
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Universiteit Gent, Ottergemsesteenweg 460, 9000 Gent, Belgium (S.V.C.)
| | - Len De Nys
- Rehabilitation Research, Vrije Universiteit Brussel (VUB), Laarbeeklaan 121, 1090 Brussels, Belgium (J.Z.); (L.D.N.)
- Medical Oncology Department, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| |
Collapse
|
38
|
Yu N, Wan Y, Zuo L, Cao Y, Qu D, Liu W, Deng L, Zhang T, Wang W, Wang J, Lv J, Xiao Z, Feng Q, Zhou Z, Bi N, Niu T, Wang X. Multi-modal radiomics features to predict overall survival of locally advanced esophageal cancer after definitive chemoradiotherapy. BMC Cancer 2025; 25:596. [PMID: 40175977 PMCID: PMC11967038 DOI: 10.1186/s12885-025-13996-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
PURPOSE To establish prediction models to predict 2-year overall survival (OS) and stratify patients with different risks based on radiomics features extracted from magnetic resonance imaging (MRI) and computed tomography (CT) before definite chemoradiotherapy (dCRT) in locally advanced esophageal squamous cell carcinoma (ESCC). METHODS Patients with locally advanced ESCC were recruited. We extracted 547 radiomics features from MRI and CT images. The least absolute shrinkage and selection operator (LASSO) for COX algorithm was used to obtain features highly correlated with survival outcomes in the training cohort. Based on MRI, CT, and the hybrid image data, three prediction models were built. The predictive performance of the radiomics models was evaluated in the training cohort and verified in the validation cohort using AUC values. RESULTS A total of 192 patients were included and randomized into the training and validation cohorts. In predicting 2-year OS, the AUCs of the CT-based model were 0.733 and 0.654 for the training and validation sets. The MRI radiomics-based model was observed with similar AUCs of 0.750 and 0.686 in the training and validation sets. The AUC values of hybrid model combining MRI and CT radiomics features in predicting 2-year OS were 0.792 and 0.715 in the training and validation cohorts. It showed significant differences in 2-year OS in the high-risk and low-risk groups divided by the best cutoff value in the hybrid radiomics-based model. CONCLUSIONS The hybrid radiomics-based model demontrated the best performance of predicting 2-year OS and can differentiate the high-risk and low-risk patients.
Collapse
Affiliation(s)
- Nuo Yu
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yidong Wan
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lijing Zuo
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Cao
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dong Qu
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenyang Liu
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Deng
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Zhang
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenqing Wang
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianyang Wang
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jima Lv
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zefen Xiao
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qinfu Feng
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zongmei Zhou
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Bi
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianye Niu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Gaoke International Innovation Center, Guangqiao Road, Guangming District, Shenzhen, China.
| | - Xin Wang
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
39
|
Wu W, Gao C, Wu L, Gao C, Li J, Su Z, Zhong H, Xu M, Sun Z. Diagnostic accuracy of deep learning for the invasiveness assessment of ground-glass nodules with fine segmentation: a systematic review and meta-analysis. Quant Imaging Med Surg 2025; 15:2722-2738. [PMID: 40235789 PMCID: PMC11994546 DOI: 10.21037/qims-24-1839] [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: 08/31/2024] [Accepted: 02/24/2025] [Indexed: 04/17/2025]
Abstract
Background Accurate recognition of invasive lung adenocarcinoma (IAC) presenting as ground-glass nodules (GGNs) is crucial for guiding clinical decision-making and timely surgical intervention. This study aimed to systematically evaluate the diagnostic accuracy of deep learning (DL) models via fine nodule segmentation in assessing the invasiveness of lung adenocarcinoma. Methods Literature from the inception of the PubMed, Embase, Cochrane Library, and Web of Science databases was searched. Studies related to DL and nodule segmentation in diagnosing IAC were evaluated and included. Titles and abstracts were screened, and the Quality Assessment of Diagnostic Accuracy Studies 2 was used to assess the quality of the selected studies. The Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) criteria of diagnostic tests were used to assess the certainty of evidence. Results Eight studies involving 5,281 nodules and 4,676 patients were included and analyzed. Meta-analysis showed that the combined sensitivity of DL for the diagnosis of IAC was 0.81 [95% confidence interval (CI): 0.73-0.87], while the specificity was 0.86 (95% CI: 0.80-0.90). The area under the summary receiver operating characteristic (SROC) curve was 0.90 (95% CI: 0.88-0.93), but the overall quality of the evidence was suboptimal. Conclusions DL and nodule segmentation demonstrated high accuracy in assessing lung adenocarcinoma invasiveness, but the certainty of the associated evidence was low. More large-scale, multicenter, high-quality diagnostic accuracy studies are needed to validate the performance and usefulness of DL in the assessment of lung adenocarcinoma invasiveness.
Collapse
Affiliation(s)
- Wei Wu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chen Gao
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Linyu Wu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuan Gao
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaying Li
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zihang Su
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haoyu Zhong
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Maosheng Xu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhichao Sun
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
40
|
Xu HL, Gong TT, Song XJ, Chen Q, Bao Q, Yao W, Xie MM, Li C, Grzegorzek M, Shi Y, Sun HZ, Li XH, Zhao YH, Gao S, Wu QJ. Artificial Intelligence Performance in Image-Based Cancer Identification: Umbrella Review of Systematic Reviews. J Med Internet Res 2025; 27:e53567. [PMID: 40167239 PMCID: PMC12000792 DOI: 10.2196/53567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 07/30/2024] [Accepted: 11/11/2024] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Artificial intelligence (AI) has the potential to transform cancer diagnosis, ultimately leading to better patient outcomes. OBJECTIVE We performed an umbrella review to summarize and critically evaluate the evidence for the AI-based imaging diagnosis of cancers. METHODS PubMed, Embase, Web of Science, Cochrane, and IEEE databases were searched for relevant systematic reviews from inception to June 19, 2024. Two independent investigators abstracted data and assessed the quality of evidence, using the Joanna Briggs Institute (JBI) Critical Appraisal Checklist for Systematic Reviews and Research Syntheses. We further assessed the quality of evidence in each meta-analysis by applying the Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) criteria. Diagnostic performance data were synthesized narratively. RESULTS In a comprehensive analysis of 158 included studies evaluating the performance of AI algorithms in noninvasive imaging diagnosis across 8 major human system cancers, the accuracy of the classifiers for central nervous system cancers varied widely (ranging from 48% to 100%). Similarities were observed in the diagnostic performance for cancers of the head and neck, respiratory system, digestive system, urinary system, female-related systems, skin, and other sites. Most meta-analyses demonstrated positive summary performance. For instance, 9 reviews meta-analyzed sensitivity and specificity for esophageal cancer, showing ranges of 90%-95% and 80%-93.8%, respectively. In the case of breast cancer detection, 8 reviews calculated the pooled sensitivity and specificity within the ranges of 75.4%-92% and 83%-90.6%, respectively. Four meta-analyses reported the ranges of sensitivity and specificity in ovarian cancer, and both were 75%-94%. Notably, in lung cancer, the pooled specificity was relatively low, primarily distributed between 65% and 80%. Furthermore, 80.4% (127/158) of the included studies were of high quality according to the JBI Critical Appraisal Checklist, with the remaining studies classified as medium quality. The GRADE assessment indicated that the overall quality of the evidence was moderate to low. CONCLUSIONS Although AI shows great potential for achieving accelerated, accurate, and more objective diagnoses of multiple cancers, there are still hurdles to overcome before its implementation in clinical settings. The present findings highlight that a concerted effort from the research community, clinicians, and policymakers is required to overcome existing hurdles and translate this potential into improved patient outcomes and health care delivery. TRIAL REGISTRATION PROSPERO CRD42022364278; https://www.crd.york.ac.uk/PROSPERO/view/CRD42022364278.
Collapse
Affiliation(s)
- He-Li Xu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ting-Ting Gong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xin-Jian Song
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qian Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qi Bao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Wei Yao
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Meng-Meng Xie
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chen Li
- Microscopic Image and Medical Image Analysis Group, College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Marcin Grzegorzek
- Institute for Medical Informatics, University of Luebeck, Luebeck, Germany
| | - Yu Shi
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hong-Zan Sun
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiao-Han Li
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu-Hong Zhao
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Song Gao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qi-Jun Wu
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China
| |
Collapse
|
41
|
Dafni MF, Shih M, Manoel AZ, Yousif MYE, Spathi S, Harshal C, Bhatt G, Chodnekar SY, Chune NS, Rasool W, Umar TP, Moustakas DC, Achkar R, Kumar H, Naz S, Acuña-Chavez LM, Evgenikos K, Gulraiz S, Ali ESM, Elaagib A, Uggh IHP. Empowering cancer prevention with AI: unlocking new frontiers in prediction, diagnosis, and intervention. Cancer Causes Control 2025; 36:353-367. [PMID: 39672997 DOI: 10.1007/s10552-024-01942-9] [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: 07/07/2024] [Accepted: 11/18/2024] [Indexed: 12/15/2024]
Abstract
Artificial intelligence is rapidly changing our world at an exponential rate and its transformative power has extensively reached important sectors like healthcare. In the fight against cancer, AI proved to be a novel and powerful tool, offering new hope for prevention and early detection. In this review, we will comprehensively explore the medical applications of AI, including early cancer detection through pathological and imaging analysis, risk stratification, patient triage, and the development of personalized prevention approaches. However, despite the successful impact AI has contributed to, we will also discuss the myriad of challenges that we have faced so far toward optimal AI implementation. There are problems when it comes to the best way in which we can use AI systemically. Having the correct data that can be understood easily must remain one of the most significant concerns in all its uses including sharing information. Another challenge that exists is how to interpret AI models because they are too complicated for people to follow through examples used in their developments which may affect trust, especially among medical professionals. Other considerations like data privacy, algorithm bias, and equitable access to AI tools have also arisen. Finally, we will evaluate possible future directions for this promising field that highlight AI's capacity to transform preventative cancer care.
Collapse
Affiliation(s)
- Marianna-Foteini Dafni
- School of Medicine, Laboratory of Forensic Medicine and Toxicology, Aristotle Univerisity of Thessaloniki, Thessaloniki, Greece
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Mohamed Shih
- School of Medicine, Newgiza University, Giza, Egypt.
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece.
| | - Agnes Zanotto Manoel
- Faculty of Medicine, Federal University of Rio Grande, Rio Grande do Sul, Brazil
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Mohamed Yousif Elamin Yousif
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Stavroula Spathi
- Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Chorya Harshal
- Faculty of Medicine, Medical College Baroda, Vadodara, India
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Gaurang Bhatt
- All India Institute of Medical Sciences, Rishikesh, India
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Swarali Yatin Chodnekar
- Faculty of Medicine, Teaching University Geomedi LLC, Tbilisi, Georgia
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Nicholas Stam Chune
- Faculty of Medicine, University of Nairobi, Nairobi, Kenya
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Warda Rasool
- Faculty of Medicine, King Edward Medical University, Lahore, Pakistan
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Tungki Pratama Umar
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Dimitrios C Moustakas
- Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Robert Achkar
- Faculty of Medicine, Poznan University of Medical Sciences, Poznan, Poland
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Harendra Kumar
- Dow University of Health Sciences, Karachi, Pakistan
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Suhaila Naz
- Tbilisi State Medical University, Tbilisi, Georgia
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Luis M Acuña-Chavez
- Facultad de Medicina de la Universidad Nacional de Trujillo, Trujillo, Peru
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Konstantinos Evgenikos
- Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Shaina Gulraiz
- Royal Bournemouth Hospital (University Hospitals Dorset), Bournemouth, UK
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Eslam Salih Musa Ali
- University of Dongola Faculty of Medicine and Health Science, Dongola, Sudan
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Amna Elaagib
- Faculty of Medicine AlMughtaribeen University, Khartoum, Sudan
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| | - Innocent H Peter Uggh
- Kilimanjaro Clinical Research Institute, Kilimanjaro, Tanzania
- Cancer Prevention Research Group in Greece, Kifisias Avenue 44, Marousi, Greece
| |
Collapse
|
42
|
Guo C, He Y, Shi Z, Wang L. Artificial intelligence in surgical medicine: a brief review. Ann Med Surg (Lond) 2025; 87:2180-2186. [PMID: 40212138 PMCID: PMC11981352 DOI: 10.1097/ms9.0000000000003115] [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: 12/13/2024] [Accepted: 02/17/2025] [Indexed: 04/13/2025] Open
Abstract
The application of artificial intelligence (AI) technology in the medical field, particularly in surgical operations, has evolved from science fiction to a crucial tool. With continuous advancements in computational power and algorithmic technology, AI is reshaping the surgical medicine landscape. From preoperative diagnosis and planning to intraoperative real-time navigation and assistance and postoperative rehabilitation and follow-up management, AI technology has significantly enhanced the precision and safety of surgical procedures. This paper systematically reviews the development and current applications of AI in surgery, focusing on specific case studies of AI in surgical procedures, diagnostic assistance, intraoperative navigation, and postoperative management, highlighting its significant contributions to improving surgical precision and safety. Despite the obvious advantages of AI in improving surgical success, reducing postoperative complications, and accelerating patient recovery, its use in surgery still faces numerous challenges, including its cost-effectiveness, dependency, data privacy and security, clinical integration, and physician training. This review summarizes the current applications of AI in surgical medicine, highlights its benefits and limitations, and discusses the challenges and future directions of integrating AI into surgical practice.
Collapse
Affiliation(s)
- Chen Guo
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yutao He
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhitian Shi
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
43
|
Wang DD, Lin S, Lyu GR. Advances in the Application of Artificial Intelligence in the Ultrasound Diagnosis of Vulnerable Carotid Atherosclerotic Plaque. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:607-614. [PMID: 39828500 DOI: 10.1016/j.ultrasmedbio.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025]
Abstract
Vulnerable atherosclerotic plaque is a type of plaque that poses a significant risk of high mortality in patients with cardiovascular disease. Ultrasound has long been used for carotid atherosclerosis screening and plaque assessment due to its safety, low cost and non-invasive nature. However, conventional ultrasound techniques have limitations such as subjectivity, operator dependence, and low inter-observer agreement, leading to inconsistent and possibly inaccurate diagnoses. In recent years, a promising approach to address these limitations has emerged through the integration of artificial intelligence (AI) into ultrasound imaging. It was found that by training AI algorithms with large data sets of ultrasound images, the technology can learn to recognize specific characteristics and patterns associated with vulnerable plaques. This allows for a more objective and consistent assessment, leading to improved diagnostic accuracy. This article reviews the application of AI in the field of diagnostic ultrasound, with a particular focus on carotid vulnerable plaques, and discusses the limitations and prospects of AI-assisted ultrasound. This review also provides a deeper understanding of the role of AI in diagnostic ultrasound and promotes more research in the field.
Collapse
Affiliation(s)
- Dan-Dan Wang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China; Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, Australia
| | - Guo-Rong Lyu
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China; Departments of Medical Imaging, Quanzhou Medical College, Quanzhou, China.
| |
Collapse
|
44
|
Li S, Hu Y, Tian C, Luan J, Zhang X, Wei Q, Li X, Bian Y. Prediction of EGFR-TP53 genes co-mutations in patients with lung adenocarcinoma (LUAD) by 18F-FDG PET/CT radiomics. Clin Transl Oncol 2025; 27:1506-1515. [PMID: 39251494 DOI: 10.1007/s12094-024-03685-0] [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: 06/16/2024] [Accepted: 08/20/2024] [Indexed: 09/11/2024]
Abstract
PURPOSE This retrospective study was undertaken to assess the predictive efficacy of 18F-FDG PET/CT -derived radiomic features concerning the co-mutation status of epidermal growth factor receptor (EGFR) and TP53 in LUAD. METHODS A cohort of 150 LUAD patients underwent pretreatment 18F-FDG PET/CT scans with known mutation status of EGFR and TP53 were collected. The feature extraction based on their PET/CT images utilized the Pyradiomics package based on the 3D Slicer. The optimal radiomic features were selected through correlation analysis and the Gradient Boosting Decision Tree (GBDT) algorithm, followed by the construction of the radiomic model. The clinical model incorporated meaningful clinical variables, whereas the complex model integrated both the radiomic and clinical models. The area under the receiver operating characteristic curve (AUC) facilitated the comparison of prediction performance across the three models. The DCA gauged the clinical utility of these models. RESULTS The patient cohort was randomly allocated into a training set (n = 105) and a validation set (n = 45) in a 7:3 ratio. Eleven PET and eleven CT optimal radiomic features were selected to construct the radiomic model. The model showed a good ability to discriminate the co-occurrence of EGFR and TP53, with AUC equal to 0.850 in the training set, and 0.748 in the validation set, compared with 0.750 and 0.626 for the clinical model. The complex model exhibited the highest AUC values, with 0.880 and 0.794 in both sets, but there were no significant differences compared to the radiomic model. The DCA revealed favorable clinical value. CONCLUSION
Collapse
Affiliation(s)
- Shuheng Li
- Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
- Department of Nuclear Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yujing Hu
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Congna Tian
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Jiusong Luan
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Xinchao Zhang
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Qiang Wei
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Xiaodong Li
- Department of Nuclear Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yanzhu Bian
- Hebei Medical University, Shijiazhuang, Hebei, China.
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China.
| |
Collapse
|
45
|
Tang J, Karbhari N, Campian JL. Therapeutic Targets in Glioblastoma: Molecular Pathways, Emerging Strategies, and Future Directions. Cells 2025; 14:494. [PMID: 40214448 PMCID: PMC11988183 DOI: 10.3390/cells14070494] [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: 02/14/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults, characterized by rapid growth, invasive infiltration into surrounding brain tissue, and resistance to conventional therapies. Despite advancements in surgery, radiotherapy, and chemotherapy, median survival remains approximately 15 months, underscoring the urgent need for innovative treatments. Key considerations informing treatment development include oncogenic genetic and epigenetic alterations that may dually serve as therapeutic targets and facilitate treatment resistance. Various immunotherapeutic strategies have been explored and continue to be refined for their anti-tumor potential. Technical aspects of drug delivery and blood-brain barrier (BBB) penetration have been addressed through novel vehicles and techniques including the incorporation of nanotechnology. Molecular profiling has emerged as an important tool to individualize treatment where applicable, and to identify patient populations with the most drug sensitivity. The goal of this review is to describe the spectrum of potential GBM therapeutic targets, and to provide an overview of key trial outcomes. Altogether, the progress of clinical and preclinical work must be critically evaluated in order to develop therapies for GBM with the strongest therapeutic efficacy.
Collapse
Affiliation(s)
- Justin Tang
- Department of Biomedical Science, University of Guelph, Guelph, ON N1G 2W1, Canada
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (N.K.); (J.L.C.)
| | - Nishika Karbhari
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (N.K.); (J.L.C.)
| | - Jian L. Campian
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (N.K.); (J.L.C.)
| |
Collapse
|
46
|
Ma S, Zhu Y, Pu C, Li J, Zhong B. Computed tomography radiomics combined with clinical parameters for hepatocellular carcinoma differentiation: a machine learning investigation. Pol J Radiol 2025; 90:e140-e150. [PMID: 40321709 PMCID: PMC12049157 DOI: 10.5114/pjr/200631] [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: 11/13/2024] [Accepted: 01/29/2025] [Indexed: 05/08/2025] Open
Abstract
Purpose To evaluate the performance of a combined clinical-radiomics model using multiple machine learning approaches for predicting pathological differentiation in hepatocellular carcinoma (HCC). Material and methods A total of 196 patients with pathologically confirmed HCC, who underwent preoperative computed tomography (CT) were retrospectively enrolled (training: n = 156; validation: n = 40). The modelling process included the folowing: (1) clinical model construction through logistic regression analysis of risk factors; (2) radiomics model development by comparing 6 machine learning classifiers; and (3) integration of optimal clinical and radiomic features into a combined model. Model performance was assessed using the area under the curve (AUC), calibration curves, and decision curve analysis (DCA). A nomogram was constructed for clinical implementation. Results Two clinical risk factors (BMI and CA153) were identified as independent predictors of differentiated HCC. The clinical model showed moderate performance (AUC: training = 0.705, validation = 0.658). The radiomics model demonstrated improved prediction capability (AUC: training = 0.840, validation = 0.716). The combined model achieved the best performance in differentiating HCC pathological grades (AUC: training = 0.878, validation = 0.747). Conclusions The integration of CT radiomics features with clinical parameters through machine learning provides a promising non-invasive approach for predicting HCC pathological differentiation. This combined model could serve as a valuable tool for preoperative treatment planning.
Collapse
Affiliation(s)
- Shijing Ma
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise City, China
- Department of Radiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise City, China
| | | | - Changhong Pu
- Department of Radiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise City, China
| | - Jin Li
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise City, China
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Bin Zhong
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise City, China
- Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, Baise City, China
| |
Collapse
|
47
|
Masroori Z, Mirghaderi P, Haseli S, Azhideh A, Mansoori B, Chen E, Park C, Chalian M. Pictorial Review of Soft Tissue Lesions with Calcification. Diagnostics (Basel) 2025; 15:811. [PMID: 40218160 PMCID: PMC11988457 DOI: 10.3390/diagnostics15070811] [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: 02/07/2025] [Revised: 02/25/2025] [Accepted: 03/01/2025] [Indexed: 04/14/2025] Open
Abstract
Calcifications in soft tissue tumors present critical diagnostic challenges in musculoskeletal imaging. Their presence and morphology can provide key clues for differentiating benign from malignant lesions, influencing both prognosis and management strategies. This pictorial review aims to explore the imaging characteristics, patterns, and implications of soft tissue calcifications, with a focus on distinguishing between benign and malignant soft tissue tumors based on the World Health Organization classification. A systematic evaluation of imaging findings in various soft tissue tumor subtypes, including adipocytic, smooth muscle, vascular, chondro-osseous, and tumors of uncertain differentiation, is presented. Additionally, non-neoplastic causes of soft tissue calcifications, such as metabolic and inflammatory conditions, are reviewed for comprehensive differential diagnosis. Our review shows that the presence, distribution, and morphology of calcifications, such as stippled, punctate, coarse, and amorphous patterns, play a crucial role in tumor characterization. Some important examples are phleboliths, which strongly suggest a benign hemangioma, while dystrophic calcification is more commonly associated with malignant entities such as synovial sarcoma and dedifferentiated liposarcoma. Peripheral calcifications with zonal distribution are characteristic of myositis ossificans, whereas central dense calcifications may indicate extra-skeletal osteosarcoma. The review also discusses the significance of calcifications in non-neoplastic conditions, such as calcific tendinitis, tumoral calcinosis, and metabolic diseases, which can mimic soft tissue tumors. Recognizing the imaging characteristics of soft tissue calcifications is essential for accurate tumor classification and appropriate clinical management. This review highlights the importance of integrating radiologic findings with clinical and histopathological data to avoid misdiagnosis and unnecessary interventions.
Collapse
Affiliation(s)
- Zahra Masroori
- Department of Radiology, Division of Musculoskeletal Imaging and Intervention, University of Washington, Seattle, WA 98105, USA (M.C.)
| | - Peyman Mirghaderi
- Department of Radiology, Division of Musculoskeletal Imaging and Intervention, University of Washington, Seattle, WA 98105, USA (M.C.)
| | - Sara Haseli
- Department of Radiology, Division of Musculoskeletal Imaging and Intervention, University of Washington, Seattle, WA 98105, USA (M.C.)
| | - Arash Azhideh
- Department of Radiology, Division of Musculoskeletal Imaging and Intervention, University of Washington, Seattle, WA 98105, USA (M.C.)
| | - Bahar Mansoori
- Department of Radiology, Division of Abdominal Imaging, University of Washington, Seattle, WA 98105, USA
| | - Eric Chen
- Department of Radiology, Division of Musculoskeletal Imaging and Intervention, University of Washington, Seattle, WA 98105, USA (M.C.)
| | - Chankue Park
- Department of Radiology, Division of Musculoskeletal Imaging and Intervention, University of Washington, Seattle, WA 98105, USA (M.C.)
| | - Majid Chalian
- Department of Radiology, Division of Musculoskeletal Imaging and Intervention, University of Washington, Seattle, WA 98105, USA (M.C.)
| |
Collapse
|
48
|
Terzi M, Yavuz MC, Bicer T, Buyuk SK. Evaluation of artificial intelligence robot's knowledge and reliability on dental implants and peri-implant phenotype. Sci Rep 2025; 15:9519. [PMID: 40108433 PMCID: PMC11923262 DOI: 10.1038/s41598-025-94576-z] [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] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/14/2025] [Indexed: 03/22/2025] Open
Abstract
The aim of this study was to evaluate the reliability and quality of information generated by ChatGPT regarding dental implants and peri-implant phenotypes. A structured questionnaire on these topics was presented to the AI-based chatbot, and its responses were assessed by dental professionals using a modified Global Quality Scale (GQS) and the DISCERN tool. The study included 60 participants divided into three professional groups: oral and maxillofacial surgeons, periodontologists, and general dental practitioners. While no statistically significant differences were observed among the groups (p > 0.05), oral and maxillofacial surgeons consistently assigned lower DISCERN and GQS scores compared to other professionals. The findings of this study suggest that ChatGPT has the potential to serve as a supplementary tool for patient information in dental implant procedures. However, its responses may lack the depth and specificity required for clinical decision-making. Dental professionals should exercise caution when relying on artificial intelligence (AI) -generated content and guide patients in interpreting such information. Future research should explore the variability of AI responses, assess multiple chatbot platforms, and investigate their integration into dental clinical practice.
Collapse
Affiliation(s)
- Mithat Terzi
- Department of Periodontology, Faculty of Dentistry, Istanbul Medeniyet University, Istanbul, Turkey
| | - Mustafa Cihan Yavuz
- Department of Periodontology, Faculty of Dentistry, Istanbul Medeniyet University, Istanbul, Turkey
| | - Tayyip Bicer
- Department of Orthodontics, Faculty of Dentistry, Ordu University, Ordu, Turkey.
| | - S Kutalmış Buyuk
- Department of Orthodontics, Faculty of Dentistry, Ordu University, Ordu, Turkey
| |
Collapse
|
49
|
Alum EU. AI-driven biomarker discovery: enhancing precision in cancer diagnosis and prognosis. Discov Oncol 2025; 16:313. [PMID: 40082367 PMCID: PMC11906928 DOI: 10.1007/s12672-025-02064-7] [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: 10/14/2024] [Accepted: 03/05/2025] [Indexed: 03/16/2025] Open
Abstract
Cancer remains a significant health issue, resulting in around 10 million deaths per year, particularly in developing nations. Demographic changes, socio-economic variables, and lifestyle choices are responsible for the rise in cancer cases. Despite the potential to mitigate the adverse effects of cancer by early detection and the implementation of cancer prevention methods, several nations have limited screening facilities. In oncology, the use of artificial intelligence (AI) represents a transformative advancement in cancer diagnosis, prognosis, and treatment. The use of AI in biomarker discovery improves precision medicine by uncovering biomarker signatures that are essential for early detection and treatment of diseases within vast and diverse datasets. Deep learning and machine learning diagnostics are two examples of AI technologies that are changing the way biomarkers are made by finding patterns in large datasets and making new technologies that make it possible to deliver accurate and effective therapies. Existing gaps include data quality, algorithmic transparency, and ethical concerns around privacy, among others. The advancement of biomarker discovery methodologies with AI seeks to transform cancer by improving patient survival rates through enhanced early diagnosis and targeted therapy. This commentary aims to clarify how AI is improving the identification of novel biomarkers for optimal early diagnosis, focused treatment, and improved clinical outcomes, while also addressing certain obstacles and ethical issues related to the application of artificial intelligence in oncology. Data from reputable scientific databases such as PubMed, Scopus, and ScienceDirect were utilized.
Collapse
Affiliation(s)
- Esther Ugo Alum
- Department of Research and Publications, Kampala International University, P. O. Box 20000, Kampala, Uganda.
| |
Collapse
|
50
|
Li Y, Sun H, Cao D, Guo Y, Wu D, Yang M, Wang H, Shao X, Li Y, Liang Y. Overcoming Biological Barriers in Cancer Therapy: Cell Membrane-Based Nanocarrier Strategies for Precision Delivery. Int J Nanomedicine 2025; 20:3113-3145. [PMID: 40098719 PMCID: PMC11913051 DOI: 10.2147/ijn.s497510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Given the unique capabilities of natural cell membranes, such as prolonged blood circulation and homotypic targeting, extensive research has been devoted to developing cell membrane-inspired nanocarriers for cancer therapy, while most focused on overcoming one or a few biological barriers. In fact, the journey of nanosystems from systemic circulation to tumor cells involves intricate processes, encompassing blood circulation, tissue accumulation, cancer cell targeting, endocytosis, endosomal escape, intracellular trafficking to target sites, and therapeutic action, all of which pose limitations to their clinical translation. This underscores the necessity of meticulously considering these biological barriers in the design of cell membrane-mimetic nanocarriers. In this review, we delineate the functions and applications of diverse types of cell membranes in nanocarrier systems. We elaborate on the biological hurdles encountered at each stage of the biomimetic nanoparticle's odyssey to the target, and comprehensively discuss the obstacles imposed by the tumor microenvironment for precise delivery. Subsequently, we systematically review contemporary cell membrane-based strategies aimed at overcoming these multi-level biological barriers, encompassing hybrid cell membrane (HCM) camouflage, tumor microenvironment remodeling, endosomal/lysosomal escape, multidrug resistance (MDR) reversal, optimization of nanoparticle physicochemical properties, and so on. Finally, we outline potential strategies to accelerate the development of cell membrane-inspired precision nanocarriers and discuss the challenges that must be addressed to enhance their clinical applicability. This review serves as a guide for refining the study of cell membrane-mimetic nanosystems in surmounting in vivo delivery barriers, thereby significantly contributing to advancing the development and application of cell membrane-based nanoparticles in cancer delivery.
Collapse
Affiliation(s)
- Yuping Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Hongfang Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Dianchao Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Yang Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Dongyang Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Menghao Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Hongming Wang
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Xiaowei Shao
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Yan Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| |
Collapse
|