1
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [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
|
2
|
Liu C, Xiu C, Zou Y, Wu W, Huang Y, Wan L, Xu S, Han B, Zhang H. Cervical cancer diagnosis model using spontaneous Raman and Coherent anti-Stokes Raman spectroscopy with artificial intelligence. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 327:125353. [PMID: 39481169 DOI: 10.1016/j.saa.2024.125353] [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: 05/08/2024] [Revised: 08/16/2024] [Accepted: 10/26/2024] [Indexed: 11/02/2024]
Abstract
Cervical cancer is the fourth most common cancer worldwide. Histopathology, which is currently considered the gold standard for cervical cancer diagnosis, can be time-consuming and subjective. Therefore, there is an urgent need for a rapid, objective, and non-destructive cervical cancer detection technique. In this study, high-wavenumber spontaneous Raman spectroscopy was used to detect cervical squamous cell carcinoma and normal tissues. The levels of lipids, fatty acids, and proteins in cervical cancerous tissues were found to be higher than those in normal tissues. Raman difference spectroscopy revealed the most significant difference at 2928 cm-1. Additionally, a Coherent anti-Stokes Raman spectroscopy (CARS) instrument was employed to enhance the wavenumber signal intensity and sensitivity. The intrinsic relationship between CARS imaging and cervical lesions was established. The CARS images indicated that the intensity of normal cervical squamous cells was zero, whereas the intensities of keratinized and non-keratinized cervical squamous cell carcinoma tissues were significantly higher. Consequently, diagnostic outcomes could be obtained by observing CARS images with the naked eye. Furthermore, the characteristic structure of keratin pearls in keratinized cervical cancer could serve as a marker for subdividing cervical cancer types. Finally, a ConvNeXt network, a machine-learning model built from CARS images, was utilized to classify different types of tissue images. The results indicated a verification accuracy of 100 %, with a loss function of 0.0927. These findings suggest that the diagnostic model established using CARS images could efficiently diagnose cervical cancer, providing novel insights into the pathological diagnosis of this disease.
Collapse
Affiliation(s)
- Chenyang Liu
- The Department of Gynecology, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Changchun 130000, China.
| | - Caifeng Xiu
- The Department of Cadre's Wards Ultrasound Diagnostics, Ultrasound Diagnostic Center, The First Hospital of Jilin University, Changchun 130000, China.
| | - Yongfang Zou
- The Department of Radiology, Changchun Infectious Disease Hospital, Changchun 130000, China.
| | - Weina Wu
- The Department of Gynecology, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Changchun 130000, China.
| | - Yizhi Huang
- The Department of Gynecology, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Changchun 130000, China.
| | - Lili Wan
- The Department of Gynecology, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Changchun 130000, China.
| | - Shuping Xu
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry of Jilin University, Changchun 130000, China.
| | - Bing Han
- The Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130000, China.
| | - Haipeng Zhang
- The Department of Gynecology, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
3
|
Chen J, Xing QC. Advancements and challenges in esophageal carcinoma prognostic models: A comprehensive review and future directions. World J Gastrointest Oncol 2025; 17:101379. [DOI: 10.4251/wjgo.v17.i2.101379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/04/2024] [Accepted: 11/22/2024] [Indexed: 01/18/2025] Open
Abstract
In this article, we comment on the article published by Yu et al. By employing LASSO regression and Cox proportional hazard models, the article identified nine significant variables affecting survival, including body mass index, Karnofsky performance status, and tumor-node-metastasis staging. We firmly concur with Yu et al regarding the vital significance of clinical prediction models (CPMs), including logistic regression and Cox regression for assessment in esophageal carcinoma (EC). However, the nomogram's limitations and the complexities of integrating genetic factors pose challenges. The integration of immunological data with advanced statistics offers new research directions. High-throughput sequencing and big data, facilitated by machine learning, have revolutionized cancer research but require substantial computational resources. The future of CPMs in EC depends on leveraging these technologies to improve predictive accuracy and clinical application, addressing the need for larger datasets, patient-reported outcomes, and regular updates for clinical relevance.
Collapse
Affiliation(s)
- Jia Chen
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan 411100, Hunan Province, China
| | - Qi-Chang Xing
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan 411100, Hunan Province, China
- The Affiliated Hospital, Hunan University, Changsha 410082, Hunan Province, China
| |
Collapse
|
4
|
Liu N, Huang Z, Chen J, Yang Y, Li Z, Liu Y, Xie Y, Wang X. Radiomics analysis of dual-energy CT-derived iodine maps for differentiating malignant from benign thyroid nodules. Med Phys 2025; 52:826-836. [PMID: 39530589 DOI: 10.1002/mp.17510] [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: 01/16/2024] [Revised: 09/19/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Many thyroid nodules are detected incidentally with the widespread use of sensitive imaging techniques; however, only a fraction of these nodules are malignant, resulting in unnecessary medical expenditures and anxiety. The major challenge is to differentiate benign thyroid nodules from malignant ones. The application of dual-energy computed tomography (DECT) and radiomics provides a new diagnostic approach. Studies applying radiomics from primary tumours on iodine maps to differentiate malignant from benign thyroid nodules are still lacking. PURPOSE To determine the ability of an iodine map-based radiomic nomogram in the venous phase for differentiating malignant thyroid nodules from benign nodules. METHODS A total of 141 patients with thyroid nodules who underwent DECT were enrolled and randomly assigned to the training and test cohorts between January 2018 and January 2019. The radiomic score (Rad-score) was derived from nine quantitative features of the iodine maps. Stepwise logistic regression analysis was used to develop radiomic, clinical and combined models. Age, normalized iodine concentration (NIC), and cyst changes were used to construct the clinical model. Receiver operating characteristic (ROC) curve analysis, sensitivity and specificity were performed to analyse the ability of the models to predict malignant thyroid nodules. Calibration analysis was used to test the fitness of the models. Decision curve analysis (DCA) and nomogram construction were also performed. RESULTS According to the clinical model, age (0.989 [0.984, 0.995]; p < 0.001), NIC (0.778 [0.640, 0.995]; p = 0.01), and cyst changes (0.617 [0.507, 0.751]; p < 0.001) were independently associated with malignant thyroid nodules. According to the combined model, age (0.994 [0.989, 0.999]; p = 0.01), NIC (0.797 [0.674, 0.941]; p = 0.008), cyst changes (0.786 [0.653, 0.947]; p = 0.01), and the rad-score (1.106 [1.070, 1.143]; p < 0.001) were independently associated with malignant thyroid nodules. The combined model achieved satisfactory discrimination in predicting malignant thyroid nodules and had greater predictive value in the training (AUC [areas under the curve], 0.96 vs. 0.87; p = 0.01) and test (AUC, 0.90 vs. 0.79; p = 0.04) cohorts than did the clinical model. CONCLUSIONS The radiomics nomogram based on iodine maps is useful to distinguish malignant thyroid nodules from benign thyroid nodules.
Collapse
Affiliation(s)
- Ni Liu
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zengfa Huang
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Chen
- Bayer Healthcare, Wuhan, Hubei, China
| | - Yang Yang
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zuoqin Li
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuanzhi Liu
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuanliang Xie
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiang Wang
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
5
|
Ahanger AB, Aalam SW, Masoodi TA, Shah A, Khan MA, Bhat AA, Assad A, Macha MA, Bhat MR. Radiogenomics and machine learning predict oncogenic signaling pathways in glioblastoma. J Transl Med 2025; 23:121. [PMID: 39871351 PMCID: PMC11773707 DOI: 10.1186/s12967-025-06101-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/08/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a highly aggressive brain tumor associated with a poor patient prognosis. The survival rate remains low despite standard therapies, highlighting the urgent need for novel treatment strategies. Advanced imaging techniques, particularly magnetic resonance imaging (MRI), are crucial in assessing GBM. Disruptions in various oncogenic signaling pathways, such as Receptor Tyrosine Kinase (RTK)-Ras-Extracellular signal-regulated kinase (ERK) signaling, Phosphoinositide 3- Kinases (PI3Ks), tumor protein p53 (TP53), and Neurogenic locus notch homolog protein (NOTCH), contribute to the development of different tumor types, each exhibiting distinct morphological and phenotypic features that can be observed at a microscopic level. However, identifying genetic abnormalities for targeted therapy often requires invasive procedures, prompting exploration into non-invasive approaches like radiogenomics. This study explores the utility of radiogenomics and machine learning (ML) in predicting these oncogenic signaling pathways in GBM patients. METHODS We collected post-operative MRI scans (T1w, T1c, FLAIR, T2w) from the BRATS-19 dataset, including scans from patients with both GBM and LGG, linked to genetic and clinical data via TCGA and CPTAC. Signaling pathway data was manually extracted from cBioPortal. Radiomic features were extracted from four MRI modalities using PyRadiomics. Dimensionality reduction and feature selection were applied and Data imbalance was addressed with SMOTE. Five ML models were trained to predict signaling pathways, with Grid Search optimizing hyperparameters and 5-fold cross-validation ensuring unbiased performance. Each model's performance was evaluated using various metrics on test data. RESULTS Our results showed a positive association between most signaling pathways and the radiomic features derived from MRI scans. The best models achieved high AUC scores, namely 0.7 for RTK-RAS, 0.8 for PI3K, 0.75 for TP53, and 0.4 for NOTCH, and therefore, demonstrated the potential of ML models in accurately predicting oncogenic signaling pathways from radiomic features, thereby informing personalized therapeutic approaches and improving patient outcomes. CONCLUSION We present a novel approach for the non-invasive prediction of deregulation in oncogenic signaling pathways in glioblastoma (GBM) by integrating radiogenomic data with machine learning models. This research contributes to advancing precision medicine in GBM management, highlighting the importance of integrating radiomics with genomic data to understand tumor behavior and treatment response better.
Collapse
Affiliation(s)
- Abdul Basit Ahanger
- Department of Computer Science, Islamic University of Science and Technology (IUST), Kashmir, 192122, India
| | - Syed Wajid Aalam
- Department of Computer Science, Islamic University of Science and Technology (IUST), Kashmir, 192122, India
| | | | - Asma Shah
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology (IUST), Kashmir, 192122, India
| | - Meraj Alam Khan
- DigiBiomics Inc, 3052 Owls Foot Drive, Mississauga, ON, Canada
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Assif Assad
- Department of Computer Science and Engineering, Islamic University of Science and Technology (IUST), Kashmir, 192122, India
| | - Muzafar Ahmad Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology (IUST), Kashmir, 192122, India.
| | - Muzafar Rasool Bhat
- Department of Computer Science, Islamic University of Science and Technology (IUST), Kashmir, 192122, India.
| |
Collapse
|
6
|
Yang L, Xie X, Zhang J, Luo C, Bu L, Wu S, Deng W, Yao Y, Zhang X, Chen H. Nonenhancing Margin and Pial Invasion in Magnetic Resonance Imaging can Predict Isocitrate Dehydrogenase Status in Glioma Patients. World Neurosurg 2025; 195:123624. [PMID: 39732457 DOI: 10.1016/j.wneu.2024.123624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND The presence of isocitrate dehydrogenase (IDH) mutations and 1p/19q codeletion significantly influences the diagnosis and prognosis of patients with lower-grade gliomas (LGGs). The ability to predict these molecular signatures preoperatively can inform surgical strategies. This study sought to establish an interpretable imaging feature set for predicting molecular signatures and overall survival in LGGs. METHODS A cohort of 113 patients with grade 2 or 3 glioma (66 with mutated IDH and 47 with wild-type IDH) was analyzed. The feature set, chief complaints, and onset symptoms were integrated into a logistic regression model to predict IDH mutation and 1p/19q codeletion statuses. Receiver operator characteristic and area under the curve analyses were performed. The predictive model was externally validated using a public database from The Cancer Genome Atlas. RESULTS Smooth nonenhancing margin and pial invasion were significant predictors of IDH mutation, with odds ratio values of 3.55 (P = 0.03) and 7.89 (P = 1.0 × 10-3), respectively. Using the Visually Accessible Rembrandt Images feature set alone to predict IDH mutation status yielded an area under the curve value of 0.83, which increased to 0.85 and 0.87 when incorporating clinical information and onset symptoms for predicting IDH mutation and 1p/19q codeletion, respectively. CONCLUSIONS Gliomas with IDH mutations were more likely to exhibit smooth nonenhancing margins and pial invasion. In clinical practice, imaging prediction allows for the assessment of IDH mutation to shift from a postoperative outcome to a preoperative guidance indicator, facilitating more precise treatment for patients with LGGs.
Collapse
Affiliation(s)
- Luhao Yang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Xian Xie
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
| | - Jie Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Chen Luo
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Linghao Bu
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shuai Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Wei Deng
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
| | - Ye Yao
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China; National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Xiaoluo Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
| | - Hong Chen
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Qian L, Fu B, He H, Liu S, Lu R. CECT-Based Radiomic Nomogram of Different Machine Learning Models for Differentiating Malignant and Benign Solid-Containing Renal Masses. J Multidiscip Healthc 2025; 18:421-433. [PMID: 39881821 PMCID: PMC11776415 DOI: 10.2147/jmdh.s502210] [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: 11/13/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
Objective This study aimed to explore the value of a radiomic nomogram based on contrast-enhanced computed tomography (CECT) for differentiating benign and malignant solid-containing renal masses. Materials and Methods A total of 122 patients with pathologically confirmed benign (n=47) or malignant (n=75) solid-containing renal masses were enrolled in this study. Radiomic features were extracted from the arterial, venous and delayed phases and further analysed by dimensionality reduction and selection. Four mainstream machine learning algorithm training models, namely, support vector machine (SVM), k-nearest neighbour (kNN), light gradient boosting (LightGBM) and logistic regression (LR), were constructed to determine the best classifier model. Univariate and multivariate analyses were used to determine the best clinical characteristics for constructing a clinical model. The radiomic and clinical signatures were integrated to construct a combined radiomic nomogram model. Receiver operating characteristic (ROC) curves and the area under the curve (AUC) were used to evaluate the performance of the radiomic nomogram, radiomic signature, and clinical model. Results Thirteen radiomic features were selected for the development of the radiomic signature. Among the various radiomic models, the LR model demonstrated superior predictive efficiency and robustness, yielding an AUC of 0.952 in the training cohort and 0.887 in the test cohort. The AUC for the clinical model was 0.854 in the training cohort and 0.747 in the test cohort. Furthermore, the radiomic nomogram, which incorporated sex, age, alcohol consumption history, and the radiomic signature, exhibited excellent discriminative performance, yielding an AUC of 0.973 in the training cohort and 0.900 in the test cohort. Conclusion The radiomic nomogram based on CECT offers a promising and noninvasive approach for distinguishing malignant from benign solid renal masses. This tool can be used to guide treatment strategies effectively and can provide valuable insights for clinicians.
Collapse
Affiliation(s)
- Lu Qian
- Department of Pathology, the First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, People’s Republic of China
| | - BinHai Fu
- Department of Nuclear Medicine, The First People’s Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, People’s Republic of China
| | - Hong He
- Department of Nuclear Medicine, The First People’s Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, People’s Republic of China
| | - Shan Liu
- Department of Pathology, the First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, People’s Republic of China
| | - RenCai Lu
- Department of Nuclear Medicine, The First People’s Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, People’s Republic of China
| |
Collapse
|
8
|
Wang Z, Qiu J, Shen X, Yang F, Liu X, Wang X, Ke N. A nomogram to preoperatively predict the aggressiveness of pancreatic neuroendocrine tumors based on CT features and 3D CT radiomic features. Abdom Radiol (NY) 2025:10.1007/s00261-024-04759-x. [PMID: 39841226 DOI: 10.1007/s00261-024-04759-x] [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: 10/07/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/23/2025]
Abstract
OBJECTIVES Combining Computed Tomography (CT) intuitive anatomical features with Three-Dimensional (3D) CT multimodal radiomic imaging features to construct a model for assessing the aggressiveness of pancreatic neuroendocrine tumors (pNETs) prior to surgery. METHODS This study involved 242 patients, randomly assigned to training (170) and validation (72) cohorts. Preoperative CT and 3D CT radiomic features were used to develop a model predicting pNETs aggressiveness. The aggressiveness of pNETs was characterized by a combination of factors including G3 grade, nodal involvement (N + status), presence of distant metastases, and/or recurrence of the disease. RESULTS Three distinct predictive models were constructed to evaluate the aggressiveness of pNETs using CT features, 3D CT radiomic features, and their combination. The combined model demonstrated the greatest predictive accuracy and clinical applicability in both the training and validation sets (AUCs (95% CIs) = 0.93 (0.90-0.97) and 0.89 (0.79-0.98), respectively). Subsequently, a nomogram was developed using the features from the combined model, displaying strong alignment between actual observations and predictions as indicated by the calibration curves. Using a nomogram score of 86.06, patients were classified into high- and low-aggressiveness groups, with the high-aggressiveness group demonstrating poorer overall survival and shorter disease-free survival. CONCLUSION This study presents a combined model incorporating CT and 3D CT radiomic features, which accurately predicts the aggressiveness of PNETs preoperatively.
Collapse
Affiliation(s)
- Ziyao Wang
- West China Hospital of Sichuan University, Chengdu, China
| | - Jiajun Qiu
- West China Hospital of Sichuan University, Chengdu, China
| | - Xiaoding Shen
- West China Hospital of Sichuan University, Chengdu, China
| | - Fan Yang
- West China Hospital of Sichuan University, Chengdu, China
| | - Xubao Liu
- West China Hospital of Sichuan University, Chengdu, China
| | - Xing Wang
- West China Hospital of Sichuan University, Chengdu, China.
| | - Nengwen Ke
- West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
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:S0301-5629(24)00467-8. [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] [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
|
10
|
Zerunian M, Polidori T, Palmeri F, Nardacci S, Del Gaudio A, Masci B, Tremamunno G, Polici M, De Santis D, Pucciarelli F, Laghi A, Caruso D. Artificial Intelligence and Radiomics in Cholangiocarcinoma: A Comprehensive Review. Diagnostics (Basel) 2025; 15:148. [PMID: 39857033 PMCID: PMC11763775 DOI: 10.3390/diagnostics15020148] [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: 11/15/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a malignant biliary system tumor and the second most common primary hepatic neoplasm, following hepatocellular carcinoma. CCA still has an extremely high unfavorable prognosis, regardless of type and location, and complete surgical resection remains the only curative therapeutic option; however, due to the underhanded onset and rapid progression of CCA, most patients present with advanced stages at first diagnosis, with only 30 to 60% of CCA patients eligible for surgery. Recent innovations in medical imaging combined with the use of radiomics and artificial intelligence (AI) can lead to improvements in the early detection, characterization, and pre-treatment staging of these tumors, guiding clinicians to make personalized therapeutic strategies. The aim of this review is to provide an overview of how radiological features of CCA can be analyzed through radiomics and with the help of AI for many different purposes, such as differential diagnosis, the prediction of lymph node metastasis, the defining of prognostic groups, and the prediction of early recurrence. The combination of radiomics with AI has immense potential. Still, its effectiveness in practice is yet to be validated by prospective multicentric studies that would allow for the development of standardized radiomics models.
Collapse
Affiliation(s)
- Marta Zerunian
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza–University of Rome, Radiology Unit–Sant’Andrea University Hospital, 00189 Rome, Italy; (T.P.); (F.P.); (S.N.); (A.D.G.); (B.M.); (G.T.); (M.P.); (D.D.S.); (F.P.); (A.L.); (D.C.)
| | - Tiziano Polidori
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza–University of Rome, Radiology Unit–Sant’Andrea University Hospital, 00189 Rome, Italy; (T.P.); (F.P.); (S.N.); (A.D.G.); (B.M.); (G.T.); (M.P.); (D.D.S.); (F.P.); (A.L.); (D.C.)
| | - Federica Palmeri
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza–University of Rome, Radiology Unit–Sant’Andrea University Hospital, 00189 Rome, Italy; (T.P.); (F.P.); (S.N.); (A.D.G.); (B.M.); (G.T.); (M.P.); (D.D.S.); (F.P.); (A.L.); (D.C.)
| | - Stefano Nardacci
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza–University of Rome, Radiology Unit–Sant’Andrea University Hospital, 00189 Rome, Italy; (T.P.); (F.P.); (S.N.); (A.D.G.); (B.M.); (G.T.); (M.P.); (D.D.S.); (F.P.); (A.L.); (D.C.)
| | - Antonella Del Gaudio
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza–University of Rome, Radiology Unit–Sant’Andrea University Hospital, 00189 Rome, Italy; (T.P.); (F.P.); (S.N.); (A.D.G.); (B.M.); (G.T.); (M.P.); (D.D.S.); (F.P.); (A.L.); (D.C.)
| | - Benedetta Masci
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza–University of Rome, Radiology Unit–Sant’Andrea University Hospital, 00189 Rome, Italy; (T.P.); (F.P.); (S.N.); (A.D.G.); (B.M.); (G.T.); (M.P.); (D.D.S.); (F.P.); (A.L.); (D.C.)
| | - Giuseppe Tremamunno
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza–University of Rome, Radiology Unit–Sant’Andrea University Hospital, 00189 Rome, Italy; (T.P.); (F.P.); (S.N.); (A.D.G.); (B.M.); (G.T.); (M.P.); (D.D.S.); (F.P.); (A.L.); (D.C.)
| | - Michela Polici
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza–University of Rome, Radiology Unit–Sant’Andrea University Hospital, 00189 Rome, Italy; (T.P.); (F.P.); (S.N.); (A.D.G.); (B.M.); (G.T.); (M.P.); (D.D.S.); (F.P.); (A.L.); (D.C.)
- PhD School in Translational Medicine and Oncology, Department of Medical and Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Domenico De Santis
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza–University of Rome, Radiology Unit–Sant’Andrea University Hospital, 00189 Rome, Italy; (T.P.); (F.P.); (S.N.); (A.D.G.); (B.M.); (G.T.); (M.P.); (D.D.S.); (F.P.); (A.L.); (D.C.)
| | - Francesco Pucciarelli
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza–University of Rome, Radiology Unit–Sant’Andrea University Hospital, 00189 Rome, Italy; (T.P.); (F.P.); (S.N.); (A.D.G.); (B.M.); (G.T.); (M.P.); (D.D.S.); (F.P.); (A.L.); (D.C.)
| | - Andrea Laghi
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza–University of Rome, Radiology Unit–Sant’Andrea University Hospital, 00189 Rome, Italy; (T.P.); (F.P.); (S.N.); (A.D.G.); (B.M.); (G.T.); (M.P.); (D.D.S.); (F.P.); (A.L.); (D.C.)
| | - Damiano Caruso
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza–University of Rome, Radiology Unit–Sant’Andrea University Hospital, 00189 Rome, Italy; (T.P.); (F.P.); (S.N.); (A.D.G.); (B.M.); (G.T.); (M.P.); (D.D.S.); (F.P.); (A.L.); (D.C.)
| |
Collapse
|
11
|
Xu M, Chen Y, Wu T, Chen Y, Zhuang W, Huang Y, Chen C. Global research trends in the application of artificial intelligence in oncology care: a bibliometric study. Front Oncol 2025; 14:1456144. [PMID: 39839779 PMCID: PMC11746057 DOI: 10.3389/fonc.2024.1456144] [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: 06/28/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
Objective To use bibliometric methods to analyze the prospects and development trends of artificial intelligence(AI) in oncology nursing from 1994 to 2024, providing guidance and reference for oncology nursing professionals and researchers. Methods The core set of the Web of Science database was searched for articles from 1994 to 2024. The R package "Bibliometrix" was used to analyze the main bibliometric features, creating a three-domain chart to display relationships among institutions, countries, and keywords. VOSviewer facilitated co-authorship analysis and its visualization was used for co- occurrence analysis. CiteSpace calculated citation bursts and keyword occurrences. Results A total of 517 articles were retrieved, representing 80 countries/regions. The United States had the highest number of publications, with 188 articles (36.4%), followed by China with 79 articles (15.3%). The top 10 institutions in terms of publication output were all U.S.-based universities or cancer research institutes, with Harvard University ranking first. Prominent research teams, such as those led by Repici, Aerts, and Almangush, have made significant contributions to studies on AI in tumor risk factor identification and symptom management. In recent years, the keywords with the highest burst strength were "model" and "human papillomavirus." The most studied tumor type was breast cancer. While Cancers published the highest number of articles, journals such as CA: A Cancer Journal for Clinicians and PLOS ONE had higher impact and citation rates. Conclusion By analyzing the volume of AI literature in oncology nursing, combined with the statistical analysis of institutions, core authors, journals, and keywords, the research hotspots and trends in the application of AI in oncology nursing over the past 30 years are revealed. AI in oncology nursing is entering a stage of rapid development, providing valuable reference for scholars and professionals in the field.
Collapse
Affiliation(s)
- Mianmian Xu
- Department of Urinary Surgery, Jinjiang Municipal Hospital, Quanzhou, China
| | - Yafang Chen
- Department of Neurology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Tianen Wu
- Department of Urinary Surgery, Jinjiang Municipal Hospital, Quanzhou, China
| | - Yuyan Chen
- Department of Urinary Surgery, Jinjiang Municipal Hospital, Quanzhou, China
| | - Wanling Zhuang
- Department of Urinary Surgery, Jinjiang Municipal Hospital, Quanzhou, China
| | - Yinhui Huang
- Department of Neurology, Jinjiang Municipal Hospital, Quanzhou, China
| | - Chuanzhen Chen
- Department of Nursing, Jinjiang Municipal Hospital, Quanzhou, China
| |
Collapse
|
12
|
Liu L, Li F, Liu X, Wang K, Zhao Z. Novel Computational and Artificial Intelligence Models in Cancer Research. Cancers (Basel) 2025; 17:116. [PMID: 39796743 PMCID: PMC11719689 DOI: 10.3390/cancers17010116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
The ICIBM 2023 marked the 11th annual conference of its kind, with the ICIBM recently becoming the official conference of the International Association for Intelligent Biology and Medicine (IAIBM), showcasing cutting-edge advancements at the intersection of computation and biomedical research [...].
Collapse
Affiliation(s)
- Li Liu
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
- Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Fuhai Li
- Institute for Informatics, Data Science and Biostatistics, Washington University in St. Louis, St. Louis, MO 63108, USA;
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Xiaoming Liu
- University of South Florida Genomics & College of Public Health, University of South Florida, Tampa, FL 33612, USA;
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
13
|
Zhao Y, Xiong S, Ren Q, Wang J, Li M, Yang L, Wu D, Tang K, Pan X, Chen F, Wang W, Jin S, Liu X, Lin G, Yao W, Cai L, Yang Y, Liu J, Wu J, Fu W, Sun K, Li F, Cheng B, Zhan S, Wang H, Yu Z, Liu X, Zhong R, Wang H, He P, Zheng Y, Liang P, Chen L, Hou T, Huang J, He B, Song J, Wu L, Hu C, He J, Yao J, Liang W. Deep learning using histological images for gene mutation prediction in lung cancer: a multicentre retrospective study. Lancet Oncol 2025; 26:136-146. [PMID: 39653054 DOI: 10.1016/s1470-2045(24)00599-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Accurate detection of driver gene mutations is crucial for treatment planning and predicting prognosis for patients with lung cancer. Conventional genomic testing requires high-quality tissue samples and is time-consuming and resource-consuming, and as a result, is not available for most patients, especially those in low-resource settings. We aimed to develop an annotation-free Deep learning-enabled artificial intelligence method to predict GEne Mutations (DeepGEM) from routinely acquired histological slides. METHODS In this multicentre retrospective study, we collected data for patients with lung cancer who had a biopsy and multigene next-generation sequencing done at 16 hospitals in China (with no restrictions on age, sex, or histology type), to form a large multicentre dataset comprising paired pathological image and multiple gene mutation information. We also included patients from The Cancer Genome Atlas (TCGA) publicly available dataset. Our developed model is an instance-level and bag-level co-supervised multiple instance learning method with label disambiguation design. We trained and initially tested the DeepGEM model on the internal dataset (patients from the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China), and further evaluated it on the external dataset (patients from the remaining 15 centres) and the public TCGA dataset. Additionally, a dataset of patients from the same medical centre as the internal dataset, but without overlap, was used to evaluate the model's generalisation ability to biopsy samples from lymph node metastases. The primary objective was the performance of the DeepGEM model in predicting gene mutations (area under the curve [AUC] and accuracy) in the four prespecified groups (ie, the hold-out internal test set, multicentre external test set, TCGA set, and lymph node metastases set). FINDINGS Assessable pathological images and multigene testing information were available for 3697 patients who had biopsy and multigene next-generation sequencing done between Jan 1, 2018, and March 31, 2022, at the 16 centres. We excluded 60 patients with low-quality images. We included 3767 images from 3637 consecutive patients (1978 [54·4%] men, 1514 [41·6%] women, 145 [4·0%] unknown; median age 60 years [IQR 52-67]), with 1716 patients in the internal dataset, 1718 patients in the external dataset, and 203 patients in the lymph node metastases dataset. The DeepGEM model showed robust performance in the internal dataset: for excisional biopsy samples, AUC values for gene mutation prediction ranged from 0·90 (95% CI 0·77-1·00) to 0·97 (0·93-1·00) and accuracy values ranged from 0·91 (0·85-0·98) to 0·97 (0·93-1·00); for aspiration biopsy samples, AUC values ranged from 0·85 (0·80-0·91) to 0·95 (0·86-1·00) and accuracy values ranged from 0·79 (0·74-0·85) to 0·99 (0·98-1·00). In the multicentre external dataset, for excisional biopsy samples, AUC values ranged from 0·80 (95% CI 0·75-0·85) to 0·91 (0·88-1·00) and accuracy values ranged from 0·79 (0·76-0·82) to 0·95 (0·93-0·96); for aspiration biopsy samples, AUC values ranged from 0·76 (0·70-0·83) to 0·87 (0·80-0·94) and accuracy values ranged from 0·76 (0·74-0·79) to 0·97 (0·96-0·98). The model also showed strong performance on the TCGA dataset (473 patients; 535 slides; AUC values ranged from 0·82 [95% CI 0·71-0·93] to 0·96 [0·91-1·00], accuracy values ranged from 0·79 [0·70-0·88] to 0·95 [0·90-1·00]). The DeepGEM model, trained on primary region biopsy samples, could be generalised to biopsy samples from lymph node metastases, with AUC values of 0·91 (95% CI 0·88-0·94) for EGFR and 0·88 (0·82-0·93) for KRAS and accuracy values of 0·85 (0·80-0·88) for EGFR and 0·95 (0·92-0·96) for KRAS and showed potential for prognostic prediction of targeted therapy. The model generated spatial gene mutation maps, indicating gene mutation spatial distribution. INTERPRETATION We developed an AI-based method that can provide an accurate, timely, and economical prediction of gene mutation and mutation spatial distribution. The method showed substantial potential as an assistive tool for guiding the clinical treatment of patients with lung cancer. FUNDING National Natural Science Foundation of China, the Science and Technology Planning Project of Guangzhou, and the National Key Research and Development Program of China. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China; AI Lab, Tencent, Shenzhen, China
| | - Shan Xiong
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China; Department of Thoracic Oncology and Surgery, Hengqin Hospital, The First Affiliated Hospital of Guangzhou Medical University, Hengqin, China
| | - Qin Ren
- AI Lab, Tencent, Shenzhen, China
| | - Jun Wang
- AI Lab, Tencent, Shenzhen, China
| | - Min Li
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Yang
- Department of Thoracic Surgery, Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, China
| | - Di Wu
- Department of Respiratory Medicine, Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, China
| | - Kejing Tang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaojie Pan
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Fengxia Chen
- Department of Thoracic Surgery, Hainan General Hospital, Haikou, China
| | - Wenxiang Wang
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shi Jin
- Department of Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Xianling Liu
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Gen Lin
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Wenxiu Yao
- Department of Oncology, University of Electronic Science and Technology of China, Sichuan Cancer Hospital and Institute & Cancer, The Second People's Hospital of Sichuan Province, Chengdu, China
| | - Linbo Cai
- Department of Oncology, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Yi Yang
- Department of Thoracic Surgery, Chengdu Third People's Hospital, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Jixian Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jingxun Wu
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Wenfan Fu
- Department of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Kai Sun
- AI Lab, Tencent, Shenzhen, China
| | - Feng Li
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Bo Cheng
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Shuting Zhan
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Haixuan Wang
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Ziwen Yu
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Xiwen Liu
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ran Zhong
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Huiting Wang
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Ping He
- Department of Pathology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongmei Zheng
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Peng Liang
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | | | - Ting Hou
- Burning Rock Biotech, Guangzhou, China
| | | | - Bing He
- AI Lab, Tencent, Shenzhen, China
| | - Jiangning Song
- Biomedicine Discovery Institute and Monash Data Futures Institute, Monash University, Melbourne, VIC, Australia
| | - Lin Wu
- Department of Thoracic Medical Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Chengping Hu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Jianxing He
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | | | - Wenhua Liang
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China; Department of Thoracic Oncology and Surgery, Hengqin Hospital, The First Affiliated Hospital of Guangzhou Medical University, Hengqin, China.
| |
Collapse
|
14
|
Zitu MM, Le TD, Duong T, Haddadan S, Garcia M, Amorrortu R, Zhao Y, Rollison DE, Thieu T. Large language models in cancer: potentials, risks, and safeguards. BJR ARTIFICIAL INTELLIGENCE 2025; 2:ubae019. [PMID: 39777117 PMCID: PMC11703354 DOI: 10.1093/bjrai/ubae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 10/26/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025]
Abstract
This review examines the use of large language models (LLMs) in cancer, analysing articles sourced from PubMed, Embase, and Ovid Medline, published between 2017 and 2024. Our search strategy included terms related to LLMs, cancer research, risks, safeguards, and ethical issues, focusing on studies that utilized text-based data. 59 articles were included in the review, categorized into 3 segments: quantitative studies on LLMs, chatbot-focused studies, and qualitative discussions on LLMs on cancer. Quantitative studies highlight LLMs' advanced capabilities in natural language processing (NLP), while chatbot-focused articles demonstrate their potential in clinical support and data management. Qualitative research underscores the broader implications of LLMs, including the risks and ethical considerations. Our findings suggest that LLMs, notably ChatGPT, have potential in data analysis, patient interaction, and personalized treatment in cancer care. However, the review identifies critical risks, including data biases and ethical challenges. We emphasize the need for regulatory oversight, targeted model development, and continuous evaluation. In conclusion, integrating LLMs in cancer research offers promising prospects but necessitates a balanced approach focusing on accuracy, ethical integrity, and data privacy. This review underscores the need for further study, encouraging responsible exploration and application of artificial intelligence in oncology.
Collapse
Affiliation(s)
- Md Muntasir Zitu
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States
| | - Tuan Dung Le
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States
| | - Thanh Duong
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States
| | - Shohreh Haddadan
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States
| | - Melany Garcia
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States
| | - Rossybelle Amorrortu
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States
| | - Yayi Zhao
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States
| | - Dana E Rollison
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States
| | - Thanh Thieu
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States
| |
Collapse
|
15
|
Andel PCM, van Goor IWJM, Augustinus S, Berrevoet F, Besselink MG, Bhojwani R, Boggi U, Bouwense SAW, Cirkel GA, van Dam JL, Djanani A, Dorcaratto D, Dreyer S, den Dulk M, Frigerio I, Ghorbani P, Goetz MR, Groot Koerkamp B, Gryspeerdt F, Hidalgo Salinas C, Intven M, Izbicki JR, Jorba Martin R, Kauffmann EF, Klug R, Liem MSL, Luyer MDP, Maglione M, Martin-Perez E, Meerdink M, de Meijer VE, Nieuwenhuijs VB, Nikov A, Nunes V, Pando Rau E, Radenkovic D, Roeyen G, Sanchez-Bueno F, Serrablo A, Sparrelid E, Tepetes K, Thakkar RG, Tzimas GN, Verdonk RC, ten Winkel M, Zerbi A, Groot VP, Molenaar IQ, Daamen LA, van Santvoort HC. Routine Imaging or Symptomatic Follow-Up After Resection of Pancreatic Adenocarcinoma. JAMA Surg 2025; 160:74-84. [PMID: 39504033 PMCID: PMC11541741 DOI: 10.1001/jamasurg.2024.5024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/29/2024] [Indexed: 11/09/2024]
Abstract
Importance International guidelines lack consistency in their recommendations regarding routine imaging in the follow-up after pancreatic resection for pancreatic ductal adenocarcinoma (PDAC). Consequently, follow-up strategies differ between centers worldwide. Objective To compare clinical outcomes, including recurrence-focused treatment and survival, in patients with PDAC recurrence who received symptomatic follow-up or routine imaging after pancreatic resection in international centers affiliated with the European-African Hepato-Pancreato-Biliary Association (E-AHPBA). Design, Setting, and Participants This was a prospective, international, cross-sectional study. Patients from a total of 33 E-AHPBA centers from 13 countries were included between 2020 and 2021. According to the predefined study protocol, patients who underwent PDAC resection and were diagnosed with disease recurrence were prospectively included. Patients were stratified according to postoperative follow-up strategy: symptomatic follow-up (ie, without routine imaging) or routine imaging. Exposures Symptomatic follow-up or routine imaging in patients who underwent PDAC resection. Main Outcomes and Measures Overall survival (OS) was estimated with Kaplan-Meier curves and compared using the log-rank test. To adjust for potential confounders, multivariable logistic regression was used to evaluate the association between follow-up strategy and recurrence-focused treatment. Multivariable Cox proportional hazard analysis was used to study the independent association between follow-up strategy and OS. Results Overall, 333 patients (mean [SD] age, 65 [11] years; 184 male [55%]) with PDAC recurrence were included. Median (IQR) follow-up at time of analysis 2 years after inclusion of the last patient was 40 (30-58) months. Of the total cohort, 98 patients (29%) received symptomatic follow-up, and 235 patients (71%) received routine imaging. OS was 23 months (95% CI, 19-29 months) vs 28 months (95% CI, 24-30 months) in the groups who received symptomatic follow-up vs routine imaging, respectively (P = .01). Routine imaging was associated with receiving recurrence-focused treatment (adjusted odds ratio, 2.57; 95% CI, 1.22-5.41; P = .01) and prolonged OS (adjusted hazard ratio, 0.75; 95% CI, 0.56-.99; P = .04). Conclusion and Relevance In this international, prospective, cross-sectional study, routine follow-up imaging after pancreatic resection for PDAC was independently associated with receiving recurrence-focused treatment and prolonged OS.
Collapse
Affiliation(s)
- Paul C. M. Andel
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
- St Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands
| | - Iris W. J. M. van Goor
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Simone Augustinus
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Frederik Berrevoet
- Department of General and HPB Surgery and Liver Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Marc G. Besselink
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Rajesh Bhojwani
- Department of Surgery, Santokba Institute of Digestive Surgical Sciences, Santokba Durlabhji Memorial Hospital, Rajasthan, India
| | - Ugo Boggi
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Stefan A. W. Bouwense
- Department of Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Surgery, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Geert A. Cirkel
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jacob L. van Dam
- Department of Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Angela Djanani
- Department of Internal Medicine, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Dimitri Dorcaratto
- Department of Surgery, Hospital Clínico, University of Valencia, Biomedical Research Institute (INCLIVA), Valencia, Spain
| | - Stephan Dreyer
- Department of Academic Surgery, Glasgow Royal Infirmary, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Marcel den Dulk
- Department of Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Surgery, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Isabella Frigerio
- Pancreatic Surgical Unit, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Poya Ghorbani
- Department of Surgery, Karolinska Institutet, Solna, Sweden
| | - Mara R. Goetz
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg, Hamburg, Germany
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Filip Gryspeerdt
- Department of General and HPB Surgery and Liver Transplantation, Ghent University Hospital, Ghent, Belgium
| | | | - Martijn Intven
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg, Hamburg, Germany
| | - Rosa Jorba Martin
- Department of Surgery, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| | - Emanuele F. Kauffmann
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Reinhold Klug
- Department of General-, Visceral- and Vascular Surgery, Community Hospital Horn, Horn, Austria
| | - Mike S. L. Liem
- Department of Surgery, Medical Spectrum Twente, Enschede, the Netherlands
| | - Misha D. P. Luyer
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
| | - Manuel Maglione
- Department of Visceral, Transplant, and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Elena Martin-Perez
- Department of General and Digestive Surgery, University Hospital La Princesa, Madrid, Spain
| | - Mark Meerdink
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Vincent E. de Meijer
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | | | - Andrej Nikov
- Department of Surgery, Military University Hospital Prague, Prague, Czech Republic
| | - Vitor Nunes
- Department of Surgery, Hospital Prof Doutor Fernando Fonseca EPE, Amadora, Portugal
| | - Elizabeth Pando Rau
- Department of Hepato-Pancreatobiliary and Transplant Surgery, Hospital Vall d’Hebron, Barcelona, Spain
- Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Dejan Radenkovic
- Clinic for Digestive Surgery, University Clinical Centra of Serbia, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Geert Roeyen
- Department of HPB, Endocrine and Transplantation Surgery, Antwerp University Hospital, Antwerp, Belgium
| | - Francisco Sanchez-Bueno
- Department of General and Digestive Surgery, Hospital Clínico Universitario “Virgen de la Arrixaca,” Murcia, Spain
| | - Alejandro Serrablo
- Department of General and Digestive Surgery, Miguel Servet University Hospital, Zaragoza, Spain
| | | | | | - Rohan G. Thakkar
- Department of Surgery, Newcastle Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - George N. Tzimas
- Department of HepatoPancreatoBiliary Surgery, Hygeia Hospital, Marousi, Greece
| | - Robert C. Verdonk
- Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Department of Gastroenterology, Utrecht, the Netherlands
| | | | - Alessandro Zerbi
- Department of Pancreatic Surgery, IRCCS Humanitas Hospital, Rozzano, and Humanitas University, Pieve Emanuele, Italy
| | - Vincent P. Groot
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| | - I. Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| | - Lois A. Daamen
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hjalmar C. van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| |
Collapse
|
16
|
Yan H, Niimi T, Matsunaga T, Fukui M, Hattori A, Takamochi K, Suzuki K. Preoperatively predicting survival outcome for clinical stage IA pure-solid non-small cell lung cancer by radiomics-based machine learning. J Thorac Cardiovasc Surg 2025; 169:254-266.e9. [PMID: 38788833 DOI: 10.1016/j.jtcvs.2024.05.010] [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: 02/13/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024]
Abstract
OBJECTIVE Clinical stage IA non-small cell lung cancer (NSCLC) showing a pure-solid appearance on computed tomography is associated with a worse prognosis. This study aimed to develop and validate machine-learning models using preoperative clinical and radiomic features to predict overall survival (OS) in clinical stage IA pure-solid NSCLC. METHODS Patients who underwent lung resection for NSCLC between January 2012 and December 2020 were reviewed. The radiomic features were extracted from the intratumoral and peritumoral regions on computed tomography. The machine-learning models were developed using random survival forest and eXtreme Gradient Boosting (XGBoost) algorithms, whereas the Cox regression model was set as a benchmark. Model performance was assessed using the integrated time-dependent area under the curve (iAUC) and validated by 5-fold cross-validation. RESULTS In total, 642 patients with clinical stage IA pure-solid NSCLC were included. Among 3748 radiomic and 34 preoperative clinical features, 42 features were selected. Both machine-learning models outperformed the Cox regression model (iAUC, 0.753; 95% confidence interval [CI], 0.629-0.829). The XGBoost model showed a better performance (iAUC, 0.832; 95% CI, 0.779-0.880) than the random survival forest model (iAUC, 0.795; 95% CI, 0.734-0.856). The XGBoost model showed an excellent survival stratification performance with a significant OS difference among the low-risk (5-year OS, 100.0%), moderate low-risk (5-year OS, 88.5%), moderate high-risk (5-year OS, 75.6%), and high-risk (5-year OS, 41.7%) groups (P < .0001). CONCLUSIONS A radiomics-based machine-learning model can preoperatively and accurately predict OS and improve survival stratification in clinical stage IA pure-solid NSCLC.
Collapse
Affiliation(s)
- Haoji Yan
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Takahiro Niimi
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Takeshi Matsunaga
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Mariko Fukui
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Aritoshi Hattori
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazuya Takamochi
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kenji Suzuki
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan.
| |
Collapse
|
17
|
Jones ML, Vijayakumar S, Nittala MR, Brunson CD. An Interdisciplinary Perspective on Improving Cancer Care in the State of Mississippi as an Example of Cancer Care Improvements in the Global South. Cureus 2025; 17:e76865. [PMID: 39758867 PMCID: PMC11698381 DOI: 10.7759/cureus.76865] [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] [Accepted: 12/24/2024] [Indexed: 01/07/2025] Open
Abstract
Cancer disparities, a critical public health issue, particularly in states such as Mississippi, where socioeconomic factors significantly influence health outcomes, require our collective attention. This paper delves into the multifaceted nature of cancer disparities through a macro-level analysis of cancer data, specifically focusing on Mississippi as a microcosm of broader national and global trends. Two key indices, the Socio-Demographic Index (SDI) and the Social Deprivation Index (SDeI), provide valuable insights. The former offers a macro-level understanding of the socioeconomic factors that shape health and cancer outcomes. The latter quantifies disadvantages in small areas, identifying regions that need scientific, policy, and administrative support. The poor health care and cancer care (CC) outcomes in Mississippi are well documented and detailed here. However, SDI and SDeI data are not yet available in Mississippi. With biological, technological, and clinical research design advancements and other new innovative strategies emerging in the past decade in CC, a 'leapfrogging' of CC outcomes in Mississippi is within our reach. To achieve this goal, an interdisciplinary approach (IDA) addressing and solving the challenges faced in Mississippi is required. The IDA team must include disciplines that can determine SDI and SDeI for Mississippi and tie those findings to successfully apply new technological advances and innovations efficiently and cost-effectively by building infrastructure and developing implementation strategies. This can serve as a pilot demonstration project that will also help other similar regions within the United States, as well as the Global South.
Collapse
Affiliation(s)
- Madison L Jones
- Medical Education, Mississippi State Medical Association, Ridgeland, USA
| | - Srinivasan Vijayakumar
- Radiotherapy and Oncology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, IND
- Radiation Oncology, University of Chicago, University of Illinois Chicago, University of California, University of Mississippi Medical Center, Ridgeland, USA
- Cancer Care, Cancer Care Advisors and Consultants LLC, Ridgeland, USA
| | - Mary R Nittala
- Radiation Oncology, University of Mississippi Medical Center, Jackson, USA
| | - Claude D Brunson
- Medical Affairs, Mississippi State Medical Association, Ridgeland, USA
- Anesthesiology, University of Mississippi Medical Center, Jackson, USA
| |
Collapse
|
18
|
Wang Y, Zhang W, Liu X, Tian L, Li W, He P, Huang S, He F, Pan X. Artificial intelligence in precision medicine for lung cancer: A bibliometric analysis. Digit Health 2025; 11:20552076241300229. [PMID: 39758259 PMCID: PMC11696962 DOI: 10.1177/20552076241300229] [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: 11/16/2023] [Accepted: 10/28/2024] [Indexed: 01/07/2025] Open
Abstract
Background The increasing body of evidence has been stimulating the application of artificial intelligence (AI) in precision medicine research for lung cancer. This trend necessitates a comprehensive overview of the growing number of publications to facilitate researchers' understanding of this field. Method The bibliometric data for the current analysis was extracted from the Web of Science Core Collection database, CiteSpace, VOSviewer ,and an online website were applied to the analysis. Results After the data were filtered, this search yielded 4062 manuscripts. And 92.27% of the papers were published from 2014 onwards. The main contributing countries were China, the United States, India, Japan, and Korea. These publications were mainly published in the following scientific disciplines, including Radiology Nuclear Medicine, Medical Imaging, Oncology, and Computer Science Notably, Li Weimin and Aerts Hugo J. W. L. stand out as leading authorities in this domain. In the keyword co-occurrence and co-citation cluster analysis of the publication, the knowledge base was divided into four clusters that are more easily understood, including screening, diagnosis, treatment, and prognosis. Conclusion This bibliometric study reveals deep learning frameworks and AI-based radiomics are receiving attention. High-quality and standardized data have the potential to revolutionize lung cancer screening and diagnosis in the era of precision medicine. However, the importance of high-quality clinical datasets, the development of new and combined AI models, and their consistent assessment for advancing research on AI applications in lung cancer are highlighted before current research can be effectively applied in clinical practice.
Collapse
Affiliation(s)
- Yuchai Wang
- Department of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Weilong Zhang
- Department of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Xiang Liu
- Department of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Li Tian
- Department of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Wenjiao Li
- Department of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Peng He
- Department of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Sheng Huang
- Department of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
- Jiuzhitang Co., Ltd, Changsha, Hunan Province, China
| | - Fuyuan He
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Xue Pan
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| |
Collapse
|
19
|
Meng X, Wang X, Zhang Z, Song L, Chen J. Recent Advancements of Nanomedicine in Breast Cancer Surgery. Int J Nanomedicine 2024; 19:14143-14169. [PMID: 39759962 PMCID: PMC11699852 DOI: 10.2147/ijn.s494364] [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: 09/03/2024] [Accepted: 11/28/2024] [Indexed: 01/07/2025] Open
Abstract
Breast cancer surgery plays a pivotal role in the multidisciplinary approaches. Surgical techniques and objectives are gradually shifting from tumor complete resection towards prolonging survival, improving cosmetic outcomes, and restoring the social and psychological well-being of patients. However, surgical treatment still faces challenges such as inadequate sensitivity in sentinel lymph node localization, the need to improve intraoperative tumor boundary localization imaging, postoperative scar healing, and the risk of recurrence, necessitating other adjunct measures for improvement. To address these challenges, specificity-optimized nanomedicines have been introduced into the surgical therapeutic landscape of breast cancer. In particular, this review involves starting with an overview of breast structure and the composition of the tumor microenvironment and then introducing the guiding principle and foundation for the design of nanomedicine. Moreover, we will take the order process of breast cancer surgery diagnosis and treatment as the starting point, and adaptively propose the roles and advantages of nanomedicine in addressing the corresponding issues. Furthermore, we also involved the prospects of utilizing advanced technological approaches. Overall, this review seeks to uncover the sophisticated design and strategies of nanomedicine from a clinical standpoint, address the challenges faced in surgical treatment, and provide insights into this subject matter.
Collapse
Affiliation(s)
- Xiangyue Meng
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xin Wang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Zhihao Zhang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Linlin Song
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Ultrasound, Laboratory of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Jie Chen
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
20
|
Gravante G, Arosio AD, Curti N, Biondi R, Berardi L, Gandolfi A, Turri-Zanoni M, Castelnuovo P, Remondini D, Bignami M. Artificial intelligence and MRI in sinonasal tumors discrimination: where do we stand? Eur Arch Otorhinolaryngol 2024:10.1007/s00405-024-09169-9. [PMID: 39719474 DOI: 10.1007/s00405-024-09169-9] [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: 10/23/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND Artificial intelligence (AI) demonstrates high potential when applied to radiomic analysis of magnetic resonance imaging (MRI) to discriminate sinonasal tumors. This can enhance diagnostic suspicion beyond visual assessment alone and prior to biopsy, leading to expedite the diagnostic timeline and the treatment planning. The aim of the present work is to evaluate the current advancements and accuracy of this technology in this domain. METHODS A systematic literature review was conducted following PRISMA guidelines. Inclusion criteria comprised studies utilizing any machine learning approach applied to MRI of patients with sinonasal tumors. For each study, comprehensive data were gathered on the MRI protocols, feature extraction techniques, and classifiers employed to develop the AI model. The performance was assessed based on accuracy and area under the curve (AUC). RESULTS Fourteen studies, published between May 2017 and August 2024, were included. These studies were categorized into three groups: those examining both benign and malignant tumors, those investigating malignant tumor subpopulations, and those focusing on benign pathologies. All studies reported an AUC greater than 0.800, achieving AUC > 0.89 and accuracy > 0.81 when incorporating clinical-radiological variables. Notably, the best discrimination performance was observed in studies utilizing combined conventional MRI sequences, including T1-weighted, contrasted T1-weighted, and T2-weighted images. CONCLUSION The application of AI and radiomics in analyzing MRI scans presents significant promise for improving the discrimination of sinonasal tumors. Integrating clinical and radiological indicators enhances model performance, suggesting that future research should focus on larger patient cohorts and diverse AI methodologies to refine diagnostic accuracy and clinical utility.
Collapse
Affiliation(s)
- Giacomo Gravante
- Division of Otorhinolaryngology, Department of Biotechnology and Life Sciences, University of Insubria, Ospedale di Circolo, Varese, Italy.
- Unit of Otorhinolaryngology, Department of Biotechnology and Life Sciences, Ospedale di Circolo e Fondazione Macchi, University of Insubria, Via Guicciardini 9, Varese, 21100, Italy.
| | - Alberto Daniele Arosio
- Division of Otorhinolaryngology, Department of Biotechnology and Life Sciences, University of Insubria, Ospedale di Circolo, Varese, Italy
| | - Nico Curti
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - Riccardo Biondi
- Data Science and Bioinformatics Laboratory, IRCCS Institute of Neurological Sciences of Bologna, Bologna, Italy
| | - Luigi Berardi
- Division of Otorhinolaryngology, Department of Biotechnology and Life Sciences, University of Insubria, Ospedale di Circolo, Varese, Italy
| | - Alberto Gandolfi
- Division of Otorhinolaryngology, Department of Biotechnology and Life Sciences, University of Insubria, Ospedale di Circolo, Varese, Italy
| | - Mario Turri-Zanoni
- Division of Otorhinolaryngology, Department of Biotechnology and Life Sciences, University of Insubria, Ospedale Sant'Anna, Como, Italy
- Head and Neck Surgery & Forensic Dissection Research Center (HNS&FDRc), Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Paolo Castelnuovo
- Division of Otorhinolaryngology, Department of Biotechnology and Life Sciences, University of Insubria, Ospedale di Circolo, Varese, Italy
- Head and Neck Surgery & Forensic Dissection Research Center (HNS&FDRc), Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Daniel Remondini
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - Maurizio Bignami
- Division of Otorhinolaryngology, Department of Biotechnology and Life Sciences, University of Insubria, Ospedale di Circolo, Varese, Italy
- Head and Neck Surgery & Forensic Dissection Research Center (HNS&FDRc), Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
21
|
Yu X, Zhou J, Wu Y, Bai Y, Meng N, Wu Q, Jin S, Liu H, Li P, Wang M. Assessment of MGMT promoter methylation status in glioblastoma using deep learning features from multi-sequence MRI of intratumoral and peritumoral regions. Cancer Imaging 2024; 24:172. [PMID: 39716317 DOI: 10.1186/s40644-024-00817-1] [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: 11/20/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024] Open
Abstract
OBJECTIVE This study aims to evaluate the effectiveness of deep learning features derived from multi-sequence magnetic resonance imaging (MRI) in determining the O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status among glioblastoma patients. METHODS Clinical, pathological, and MRI data of 356 glioblastoma patients (251 methylated, 105 unmethylated) were retrospectively examined from the public dataset The Cancer Imaging Archive. Each patient underwent preoperative multi-sequence brain MRI scans, which included T1-weighted imaging (T1WI) and contrast-enhanced T1-weighted imaging (CE-T1WI). Regions of interest (ROIs) were delineated to identify the necrotic tumor core (NCR), enhancing tumor (ET), and peritumoral edema (PED). The ET and NCR regions were categorized as intratumoral ROIs, whereas the PED region was categorized as peritumoral ROIs. Predictive models were developed using the Transformer algorithm based on intratumoral, peritumoral, and combined MRI features. The area under the receiver operating characteristic curve (AUC) was employed to assess predictive performance. RESULTS The ROI-based models of intratumoral and peritumoral regions, utilizing deep learning algorithms on multi-sequence MRI, were capable of predicting MGMT promoter methylation status in glioblastoma patients. The combined model of intratumoral and peritumoral regions exhibited superior diagnostic performance relative to individual models, achieving an AUC of 0.923 (95% confidence interval [CI]: 0.890 - 0.948) in stratified cross-validation, with sensitivity and specificity of 86.45% and 87.62%, respectively. CONCLUSION The deep learning model based on MRI data can effectively distinguish between glioblastoma patients with and without MGMT promoter methylation.
Collapse
Affiliation(s)
- Xuan Yu
- Department of Radiology, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450000, PR China
| | - Jing Zhou
- Department of Radiology, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450000, PR China
| | - Yaping Wu
- Department of Radiology, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450000, PR China
- Biomedical Research Institute, Henan Academy of Sciences, Zhengzhou, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, Zhengzhou, China
| | - Yan Bai
- Department of Radiology, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450000, PR China
- Biomedical Research Institute, Henan Academy of Sciences, Zhengzhou, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, Zhengzhou, China
| | - Nan Meng
- Department of Radiology, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450000, PR China
| | - Qingxia Wu
- Department of Radiology, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450000, PR China
| | - Shuting Jin
- School of Computer Science and Technology, Wuhan University of Science and Technology, Wuhan, China
| | - Huanhuan Liu
- Department of Radiology, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450000, PR China
| | - Panlong Li
- Department of Radiology, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450000, PR China
| | - Meiyun Wang
- Department of Radiology, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450000, PR China.
- Biomedical Research Institute, Henan Academy of Sciences, Zhengzhou, China.
| |
Collapse
|
22
|
Shah SAH, Shah STH, Khaled R, Buccoliero A, Shah SBH, Di Terlizzi A, Di Benedetto G, Deriu MA. Explainable AI-Based Skin Cancer Detection Using CNN, Particle Swarm Optimization and Machine Learning. J Imaging 2024; 10:332. [PMID: 39728229 PMCID: PMC11727770 DOI: 10.3390/jimaging10120332] [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: 11/25/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024] Open
Abstract
Skin cancer is among the most prevalent cancers globally, emphasizing the need for early detection and accurate diagnosis to improve outcomes. Traditional diagnostic methods, based on visual examination, are subjective, time-intensive, and require specialized expertise. Current artificial intelligence (AI) approaches for skin cancer detection face challenges such as computational inefficiency, lack of interpretability, and reliance on standalone CNN architectures. To address these limitations, this study proposes a comprehensive pipeline combining transfer learning, feature selection, and machine-learning algorithms to improve detection accuracy. Multiple pretrained CNN models were evaluated, with Xception emerging as the optimal choice for its balance of computational efficiency and performance. An ablation study further validated the effectiveness of freezing task-specific layers within the Xception architecture. Feature dimensionality was optimized using Particle Swarm Optimization, reducing dimensions from 1024 to 508, significantly enhancing computational efficiency. Machine-learning classifiers, including Subspace KNN and Medium Gaussian SVM, further improved classification accuracy. Evaluated on the ISIC 2018 and HAM10000 datasets, the proposed pipeline achieved impressive accuracies of 98.5% and 86.1%, respectively. Moreover, Explainable-AI (XAI) techniques, such as Grad-CAM, LIME, and Occlusion Sensitivity, enhanced interpretability. This approach provides a robust, efficient, and interpretable solution for automated skin cancer diagnosis in clinical applications.
Collapse
Affiliation(s)
- Syed Adil Hussain Shah
- Department of Research and Development (R&D), GPI SpA, 38123 Trento, Italy; (S.A.H.S.); (A.B.); (A.D.T.)
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Turin, Italy
| | - Syed Taimoor Hussain Shah
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Turin, Italy
| | - Roa’a Khaled
- Department of Computer Engineering, University of Cádiz, 11519 Puerto Real, Spain;
| | - Andrea Buccoliero
- Department of Research and Development (R&D), GPI SpA, 38123 Trento, Italy; (S.A.H.S.); (A.B.); (A.D.T.)
- Human Science Department, Università degli studi di Verona, Lungadige Porta Vittoria, 17, 37129 Verona, Italy
| | - Syed Baqir Hussain Shah
- Department of Computer Science, COMSATS University Islamabad (CUI), Wah Campus, Wah 47000, Pakistan;
| | - Angelo Di Terlizzi
- Department of Research and Development (R&D), GPI SpA, 38123 Trento, Italy; (S.A.H.S.); (A.B.); (A.D.T.)
| | | | - Marco Agostino Deriu
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Turin, Italy
| |
Collapse
|
23
|
Hsu CX, Chou HC, Yen SH. Exploring emergency presentations management in lung cancer: insights on oncology emergencies and histological heterogeneity. Am J Emerg Med 2024:S0735-6757(24)00731-9. [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
|
24
|
Li Y, Li W, Xiao H, Chen W, Lu J, Huang N, Li Q, Zhou K, Kojima I, Liu Y, Ou Y. Automated classification of pathological differentiation in head and neck squamous cell carcinoma using combined radiomics models from CET1WI and T2WI. Clin Oral Investig 2024; 29:25. [PMID: 39708187 DOI: 10.1007/s00784-024-06110-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVES This study aims to develop an automated radiomics-based model to grade the pathological differentiation of head and neck squamous cell carcinoma (HNSCC) and to assess the influence of various magnetic resonance imaging (MRI) sequences on the model's performance. MATERIALS AND METHODS We retrospectively analyzed MRI data from 256 patients across two medical centers, including both contrast-enhanced T1-weighted images (CET1WI) and T2-weighted images (T2WI). Regions of interest were delineated for radiomics feature extraction, followed by dimensionality reduction. An XGBoost classifier was then employed to build the predictive model, with its classification efficiency assessed using receiver operating characteristic curves and the area under the curve (AUC). RESULTS In validation cohort, the AUC (macro/micro) values for models utilizing CET1WI, T2WI, and the combination of CET1WI and T2WI were 0.801/0.814, 0.741/0.798, and 0.885/0.895, respectively. The AUC for the three differentiations, ranging from well-differentiated to poorly differentiated, were 0.867, 0.909, and 0.837, respectively. The macro/micro precision, recall, and F1 scores of 0.688/0.736, 0.744/0.828, and 0.685/0.779 for the CET1WI + T2WI model. CONCLUSION This study demonstrates that constructing a radiomics model based on CET1WI and T2WI sequences can be used to predict the pathological differentiation grading of HNSCC patients. CLINICAL RELEVANCE This study suggests that a radiomics model integrating CET1WI and T2WI MRI sequences can effectively predict the pathological differentiation of HNSCC, providing an alternative diagnostic approach through non-invasive preoperative methods.
Collapse
Affiliation(s)
- Yang Li
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Wen Li
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Haotian Xiao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weizhong Chen
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jie Lu
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Nengwen Huang
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Qingling Li
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Kangwei Zhou
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ikuho Kojima
- Department of Oral Diagnosis, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Yiming Liu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanjing Ou
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
25
|
Alqahtani MM, Alanazi AMM, Algarni SS, Aljohani H, Alenezi FK, F Alotaibi T, Alotaibi M, K Alqahtani M, Alahmari M, S Alwadeai K, M Alghamdi S, Almeshari MA, Alshammari TF, Mumenah N, Al Harbi E, Al Nufaiei ZF, Alhuthail E, Alzahrani E, Alahmadi H, Alarifi A, Zaidan A, T Ismaeil T. Unveiling the Influence of AI on Advancements in Respiratory Care: Narrative Review. Interact J Med Res 2024; 13:e57271. [PMID: 39705080 PMCID: PMC11699506 DOI: 10.2196/57271] [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: 02/10/2024] [Revised: 09/22/2024] [Accepted: 10/28/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Artificial intelligence is experiencing rapid growth, with continual innovation and advancements in the health care field. OBJECTIVE This study aims to evaluate the application of artificial intelligence technologies across various domains of respiratory care. METHODS We conducted a narrative review to examine the latest advancements in the use of artificial intelligence in the field of respiratory care. The search was independently conducted by respiratory care experts, each focusing on their respective scope of practice and area of interest. RESULTS This review illuminates the diverse applications of artificial intelligence, highlighting its use in areas associated with respiratory care. Artificial intelligence is harnessed across various areas in this field, including pulmonary diagnostics, respiratory care research, critical care or mechanical ventilation, pulmonary rehabilitation, telehealth, public health or health promotion, sleep clinics, home care, smoking or vaping behavior, and neonates and pediatrics. With its multifaceted utility, artificial intelligence can enhance the field of respiratory care, potentially leading to superior health outcomes for individuals under this extensive umbrella. CONCLUSIONS As artificial intelligence advances, elevating academic standards in the respiratory care profession becomes imperative, allowing practitioners to contribute to research and understand artificial intelligence's impact on respiratory care. The permanent integration of artificial intelligence into respiratory care creates the need for respiratory therapists to positively influence its progression. By participating in artificial intelligence development, respiratory therapists can augment their clinical capabilities, knowledge, and patient outcomes.
Collapse
Affiliation(s)
- Mohammed M Alqahtani
- Department of Respiratory Therapy, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Respiratory Services, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Abdullah M M Alanazi
- Department of Respiratory Therapy, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Respiratory Services, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Saleh S Algarni
- Department of Respiratory Therapy, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Respiratory Services, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Hassan Aljohani
- Department of Respiratory Therapy, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Respiratory Services, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Faraj K Alenezi
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Anesthesia Technology Department, College of Applied Medical Sciences, King Saud Bin Abdul-Aziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Tareq F Alotaibi
- Department of Respiratory Therapy, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Respiratory Services, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Mansour Alotaibi
- Department of Physical Therapy, Northern Border University, Arar, Saudi Arabia
| | - Mobarak K Alqahtani
- Department of Respiratory Therapy, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Respiratory Services, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Mushabbab Alahmari
- Department of Respiratory Therapy, College of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
- Health and Humanities Research Center, University of Bisha, Bisha, Saudi Arabia
| | - Khalid S Alwadeai
- Department of Rehabilitation Science, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Saeed M Alghamdi
- Clinical Technology Department, Respiratory Care Program, Faculty of Applied Medical Sciences, Umm Al-Qura University, Mekkah, Saudi Arabia
| | - Mohammed A Almeshari
- Department of Rehabilitation Science, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | | | - Noora Mumenah
- Department of Respiratory Therapy, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Respiratory Services, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Ebtihal Al Harbi
- Department of Respiratory Therapy, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Respiratory Services, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Ziyad F Al Nufaiei
- Department of Respiratory Therapy, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Eyas Alhuthail
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Basic Sciences Department, College of Sciences and Health Professions, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Esam Alzahrani
- Department of Computer Engineering, Al-Baha University, Alaqiq, Saudi Arabia
| | - Husam Alahmadi
- Department of Respiratory Therapy, Faculty of Medical Rehabilitation Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulaziz Alarifi
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Basic Sciences Department, College of Sciences and Health Professions, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Amal Zaidan
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Public Health, College of Public Health and Health Informatics, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Taha T Ismaeil
- Department of Respiratory Therapy, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Respiratory Services, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
26
|
Singh A, Singh A, Bhattacharya S. Research trends on AI in breast cancer diagnosis, and treatment over two decades. Discov Oncol 2024; 15:772. [PMID: 39692996 DOI: 10.1007/s12672-024-01671-0] [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: 07/22/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
OBJECTIVE Recently, the integration of Artificial Intelligence (AI) has significantly enhanced the diagnostic accuracy in breast cancer screening. This study aims to deliver an extensive review of the advancements in AI for breast cancer diagnosis and prognosis through a bibliometric analysis. METHODOLOGY Therefore, this study gathered pertinent peer-reviewed research articles from the Scopus database, spanning the years 2000 to 2024. These articles were subsequently subjected to quantitative analysis and visualization through the Bibliometrix R package. Ultimately, potential areas for future research challenges were pinpointed. RESULTS This study analyzes the development of Artificial Intelligence (AI) research for breast cancer diagnosis and prognosis from 2000 to 2024, based on 2678 publications sourced from Scopus. A sharp rise in global publication trends is observed between 2018 and 2023, with 2023 producing 456 papers, indicating intensified academic focus. Leading contributors include ZHENG B, with 36 publications, and institutions like RADBOUD UNIVERSITY MEDICAL CENTER and the IEO EUROPEAN INSTITUTE OF ONCOLOGY IRCCS. The USA leads both in publications (473) and total citations (18,530), followed by India with 289 papers. Co-occurrence analysis shows that "mammography" (3171 occurrences) and "artificial intelligence" (1691 occurrences) are among the most frequent keywords, reflecting core themes. Co-citation network analysis identifies foundational works by authors like Lecun Y. and Simonyan K. in advancing AI applications in breast cancer. Institutional and country-level collaboration analysis reveals the USA's significant partnerships with China, the UK, and Canada, driving the global research agenda in this field. CONCLUSION In conclusion, this bibliometric review underscores the growing influence of AI, particularly deep learning, in breast cancer diagnosis and treatment research from 2000 to 2024. The United States leads the field in publications and collaborations, with India, Spain, and the Netherlands also making significant contributions. Key institutions and journals have driven advancements, with AI applications focusing on improving diagnostic imaging and early detection. However, challenges like data limitations, regulatory hurdles, and unequal global collaboration persist, requiring further interdisciplinary efforts to enhance AI integration in clinical practice.
Collapse
Affiliation(s)
- Alok Singh
- Department of Community Medicine, Shree Guru Gobind Singh Tricentenary (SGT) University, Gurugram, Haryana, India
| | - Akanksha Singh
- Mahatma Gandhi Kashi Vidyapith (MGKV), Varanasi, Uttar Pradesh, India
| | - Sudip Bhattacharya
- Department of Community and Family Medicine, All India Institute of Medical Sciences (AIIMS), Jharkhand, Deoghar, India.
| |
Collapse
|
27
|
Shahzadi M, Rafique H, Waheed A, Naz H, Waheed A, Zokirova FR, Khan H. Artificial intelligence for chimeric antigen receptor-based therapies: a comprehensive review of current applications and future perspectives. Ther Adv Vaccines Immunother 2024; 12:25151355241305856. [PMID: 39691280 PMCID: PMC11650588 DOI: 10.1177/25151355241305856] [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: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Using artificial intelligence (AI) to enhance chimeric antigen receptor (CAR)-based therapies' design, production, and delivery is a novel and promising approach. This review provides an overview of the current applications and challenges of AI for CAR-based therapies and suggests some directions for future research and development. This paper examines some of the recent advances of AI for CAR-based therapies, for example, using deep learning (DL) to design CARs that target multiple antigens and avoid antigen escape; using natural language processing to extract relevant information from clinical reports and literature; using computer vision to analyze the morphology and phenotype of CAR cells; using reinforcement learning to optimize the dose and schedule of CAR infusion; and using AI to predict the efficacy and toxicity of CAR-based therapies. These applications demonstrate the potential of AI to improve the quality and efficiency of CAR-based therapies and to provide personalized and precise treatments for cancer patients. However, there are also some challenges and limitations of using AI for CAR-based therapies, for example, the lack of high-quality and standardized data; the need for validation and verification of AI models; the risk of bias and error in AI outputs; the ethical, legal, and social issues of using AI for health care; and the possible impact of AI on the human role and responsibility in cancer immunotherapy. It is important to establish a multidisciplinary collaboration among researchers, clinicians, regulators, and patients to address these challenges and to ensure the safe and responsible use of AI for CAR-based therapies.
Collapse
Affiliation(s)
- Muqadas Shahzadi
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Hamad Rafique
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Ahmad Waheed
- Department of Zoology, Faculty of Life Sciences, University of Okara, 2 KM Lahore Road, Renala Khurd, Okara 56130, Punjab, Pakistan
| | - Hina Naz
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Atifa Waheed
- Department of Biology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | | | - Humera Khan
- Department of Biochemistry, Sahiwal Medical College, Sahiwal, Pakistan
| |
Collapse
|
28
|
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 2024:10.1007/s10552-024-01942-9. [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] [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
|
29
|
Lee H, Chung JW, Kim KO, Kwon KA, Kim JH, Yun SC, Jung SW, Sheeraz A, Yoon YJ, Kim JH, Kayasseh MA. Validation of Artificial Intelligence Computer-Aided Detection of Colonic Neoplasm in Colonoscopy. Diagnostics (Basel) 2024; 14:2762. [PMID: 39682670 DOI: 10.3390/diagnostics14232762] [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: 10/14/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Controlling colonoscopic quality is important in the detection of colon polyps during colonoscopy as it reduces the overall long-term colorectal cancer risk. Artificial intelligence has recently been introduced in various medical fields. In this study, we aimed to validate a previously developed artificial intelligence (AI) computer-aided detection (CADe) algorithm called ALPHAON® and compare outcomes with previous studies that showed that AI outperformed and assisted endoscopists of diverse levels of expertise in detecting colon polyps. METHODS We used the retrospective data of 500 still images, including 100 polyp images and 400 healthy colon images. In addition, we validated the CADe algorithm and compared its diagnostic performance with that of two expert endoscopists and six trainees from Gachon University Gil Medical Center. After a washing-out period of over 2 weeks, endoscopists performed polyp detection on the same dataset with the assistance of ALPHAON®. RESULTS The CADe algorithm presented a high capability in detecting colon polyps, with an accuracy of 0.97 (95% CI: 0.96 to 0.99), sensitivity of 0.91 (95% CI: 0.85 to 0.97), specificity of 0.99 (95% CI: 0.97 to 0.99), and AUC of 0.967. When evaluating and comparing the polyp detection ability of ALPHAON® with that of endoscopists with different levels of expertise (regarding years of endoscopic experience), it was found that ALPHAON® outperformed the experts in accuracy (0.97, 95% CI: 0.96 to 0.99), sensitivity (0.91, 95% CI: 0.85 to 0.97), and specificity (0.99, 95% CI: 0.97 to 0.99). After a washing-out period of over 2 weeks, the overall capability significantly improved for both experts and trainees with the assistance of ALPHAON®. CONCLUSIONS The high performance of the CADe algorithm system in colon polyp detection during colonoscopy was verified. The sensitivity of ALPHAON® led to it outperforming the experts, and it demonstrated the ability to enhance the polyp detection ability of both experts and trainees, which suggests a significant possibility of ALPHAON® being able to provide endoscopic assistance.
Collapse
Affiliation(s)
- Hannah Lee
- Division of Gastroenterology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon 21565, Republic of Korea
| | - Jun-Won Chung
- Division of Gastroenterology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon 21565, Republic of Korea
| | - Kyoung Oh Kim
- Division of Gastroenterology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon 21565, Republic of Korea
| | - Kwang An Kwon
- Division of Gastroenterology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon 21565, Republic of Korea
| | - Jung Ho Kim
- Division of Gastroenterology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon 21565, Republic of Korea
| | - Sung-Cheol Yun
- Division of Biostatistics, Center for Medical Research and Information, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Sung Woo Jung
- Division of Gastroenterology, Department of Internal Medicine, Korea University College of Medicine, Ansan 15355, Republic of Korea
| | | | | | - Ji Hee Kim
- CAIMI Co., Ltd., Incheon 22004, Republic of Korea
| | - Mohd Azzam Kayasseh
- Division of Gastroenterology, Dr. Sulaiman AI Habib Medical Group, Dubai Healthcare City, Dubai 51431, United Arab Emirates
| |
Collapse
|
30
|
Begal J, Sabo E, Goldberg N, Bitterman A, Khoury W. Wavelets-Based Texture Analysis of Post Neoadjuvant Chemoradiotherapy Magnetic Resonance Imaging as a Tool for Recognition of Pathological Complete Response in Rectal Cancer, a Retrospective Study. J Clin Med 2024; 13:7383. [PMID: 39685841 DOI: 10.3390/jcm13237383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Patients with locally advanced rectal cancer (LARC) treated by neoadjuvant chemoradiotherapy (nCRT) may experience pathological complete response (pCR). Tools that can identify pCR are required to define candidates suitable for the watch and wait (WW) strategy. Automated image analysis is used for predicting clinical aspects of diseases. Texture analysis of magnetic resonance imaging (MRI) wavelets algorithms provides a novel way to identify pCR. We aimed to evaluate wavelets-based image analysis of MRI for predicting pCR. Methods: MRI images of rectal cancer from 22 patients who underwent nCRT were captured at best representative views of the tumor. The MRI images were digitized and their texture was analyzed using different mother wavelets. Each mother wavelet was used to scan the image repeatedly at different frequencies. Based on these analyses, coefficients of similarity were calculated providing a variety of textural variables that were subsequently correlated with histopathology in each case. This allowed for proper identification of the best mother wavelets able to predict pCR. The predictive formula of complete response was computed using the independent statistical variables that were singled out by the multivariate regression model. Results: The statistical model used four wavelet variables to predict pCR with an accuracy of 100%, sensitivity of 100%, specificity of 100%, and PPV and NPV of 100%. Conclusions: Wavelet-transformed texture analysis of radiomic MRI can predict pCR in patients with LARC. It may provide a potential accurate surrogate method for the prediction of clinical outcomes of nCRT, resulting in an effective selection of patients amenable to WW.
Collapse
Affiliation(s)
- Julia Begal
- Department of General Surgery, Carmel Medical Center, Haifa 3436212, Israel
| | - Edmond Sabo
- Department of Human Pathology, Carmel Medical Center, Haifa 3436212, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Natalia Goldberg
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
- Department of Radiology, Carmel Medical Center, Haifa 3436212, Israel
| | - Arie Bitterman
- Department of General Surgery, Carmel Medical Center, Haifa 3436212, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Wissam Khoury
- Department of General Surgery, Carmel Medical Center, Haifa 3436212, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
31
|
Dong F, Li J, Wang J, Yang X. Diagnostic performance of DCE-MRI radiomics in predicting axillary lymph node metastasis in breast cancer patients: A meta-analysis. PLoS One 2024; 19:e0314653. [PMID: 39625963 PMCID: PMC11614294 DOI: 10.1371/journal.pone.0314653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/13/2024] [Indexed: 12/06/2024] Open
Abstract
Radiomics offers a novel strategy for the differential diagnosis, prognosis evaluation, and prediction of treatment responses in breast cancer. Studies have explored radiomic signatures from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) for predicting axillary lymph node metastasis (ALNM) and sentinel lymph node metastasis (SLNM), but the diagnostic accuracy varies widely. To evaluate this performance, we conducted a meta-analysis performing a comprehensive literature search across databases including PubMed, EMBASE, SCOPUS, Web of Science (WOS), Cochrane Library, China National Knowledge Infrastructure (CNKI), Wanfang Data, and the Chinese BioMedical Literature Database (CBM) until March 31, 2024. The pooled sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and the area under the receiver operating characteristic curve (AUC) were calculated. Twenty-four eligible studies encompassing 5588 breast cancer patients were included in the meta-analysis. The meta-analysis yielded a pooled sensitivity of 0.81 (95% confidence interval [CI]: 0.77-0.84), specificity of 0.85 (95%CI: 0.81-0.87), PLR of 5.24 (95%CI: 4.32-6.34), NLR of 0.23 (95%CI: 0.19-0.27), DOR of 23.16 (95%CI: 17.20-31.19), and AUC of 0.90 (95%CI: 0.87-0.92), indicating good diagnostic performance. Significant heterogeneity was observed in analyses of sensitivity (I2 = 74.64%) and specificity (I2 = 83.18%). Spearman's correlation coefficient suggested no significant threshold effect (P = 0.538). Meta-regression and subgroup analyses identified several potential heterogeneity sources, including data source, integration of clinical factors and peritumor features, MRI equipment, magnetic field strength, lesion segmentation, and modeling methods. In conclusion, DCE-MRI radiomic models exhibit good diagnostic performance in predicting ALNM and SLNM in breast cancer. This non-invasive and effective tool holds potential for the preoperative diagnosis of lymph node metastasis in breast cancer patients.
Collapse
Affiliation(s)
- Fei Dong
- Department of Medical Imaging, Yuncheng Central Hospital Affiliated to Shanxi Medical University, Yuncheng, Shanxi Province, China
| | - Jie Li
- Department of Anesthesiology, Yuncheng Central Hospital Affiliated to Shanxi Medical University, Yuncheng, Shanxi Province, China
| | - Junbo Wang
- Department of Medical Imaging, Yuncheng Central Hospital Affiliated to Shanxi Medical University, Yuncheng, Shanxi Province, China
| | - Xiaohui Yang
- Department of Medical Imaging, Yuncheng Central Hospital Affiliated to Shanxi Medical University, Yuncheng, Shanxi Province, China
| |
Collapse
|
32
|
Sun C, Jiang C, Wang X, Ma S, Zhang D, Jia W. MR-Based Radiomics Predicts CDK6 Expression and Prognostic Value in High-grade Glioma. Acad Radiol 2024; 31:5141-5153. [PMID: 38964985 DOI: 10.1016/j.acra.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024]
Abstract
RATIONALE AND OBJECTIVES This study aims to assess the prognostic value of Cyclin-dependent kinases 6 (CDK6) expression levels and establish a machine learning-based radiomics model for predicting the expression levels of CDK6 in high-grade gliomas (HGG). MATERIALS AND METHODS Clinical parameters and genomic data were extracted from 310 HGG patients in the Cancer Genome Atlas (TCGA) database and 27 patients in the Repository of Molecular Brain Neoplasia Data (REMBRANDT) database. Univariate and multivariate Cox regression, as well as Kaplan-Meier analysis, were performed for prognosis analysis. The correlation between immune cell Infiltration with CDK6 was assessed using spearman correlation analysis. Radiomic features were extracted from contrast-enhanced magnetic resonance imaging (CE-MRI) in the Cancer Imaging Archive (TCIA) database (n = 82) and REMBRANDT database (n = 27). Logistic regression (LR) and support vector machine (SVM) were employed to establish the radiomics model for predicting CDK6 expression. Receiver operating characteristic curve (ROC), calibration curve, and decision curve analysis (DCA) were utilized to assess the predictive performance of the radiomics model. Generate radiomic scores (RS) based on the LR model. An RS-based nomogram was constructed to predict the prognosis of HGG. RESULTS CDK6 was significantly overexpressed in HGG tissues and was related to lower overall survival. A significant elevation in infiltrating M0 macrophages was observed in the CDK6 high group (P < 0.001). The LR radiomics model for the prediction of CDK6 expression levels (AUC=0.810 in the training cohort, AUC = 0.784 after cross-validation, AUC=0.750 in the testing cohort) was established utilizing three radiomic features. The predictive efficiencies of the RS-based nomogram, as measured by AUC, were 0.769 for 1-year, 0.815 for 3-year, and 0.780 for 5-year, respectively. CONCLUSION The expression level of CDK6 can impact the prognosis of patients with HGG. The expression level of HGG can be noninvasively prognosticated utilizing a radiomics model.
Collapse
Affiliation(s)
- Chen Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Chenggang Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Xi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Shunchang Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Dainan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China.
| |
Collapse
|
33
|
Yang Y, Yuan Z, Ren Q, Wang J, Guan S, Tang X, Jiang Q, Meng X. Machine Learning-Enabled Fuhrman Grade in Clear-cell Renal Carcinoma Prediction Using Two-dimensional Ultrasound Images. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1911-1918. [PMID: 39317624 DOI: 10.1016/j.ultrasmedbio.2024.08.019] [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: 04/04/2024] [Revised: 08/22/2024] [Accepted: 08/24/2024] [Indexed: 09/26/2024]
Abstract
OBJECTIVE Accurate assessment of Fuhrman grade is crucial for optimal clinical management and personalized treatment strategies in patients with clear cell renal cell carcinoma (CCRCC). In this study, we developed a predictive model using ultrasound (US) images to accurately predict the Fuhrman grade. METHODS Between March 2013 and July 2023, a retrospective analysis was conducted on the US imaging and clinical data of 235 patients with pathologically confirmed CCRCC, including 67 with Fuhrman grades Ⅲ and Ⅳ. This study included 201 patients from Hospital A who were divided into training set (n = 161) and an internal validation set (n = 40) in an 8:2 ratio. Additionally, 34 patients from Hospital B were included for external validation. US images were delineated using ITK software, and radiomics features were extracted using PyRadiomics software. Subsequently, separate models for clinical factors, radiomics features, and their combinations were constructed. The model's performance was assessed by calculating the area under the receiver operating characteristic curve (AUC), calibration curve and decision curve analysis (DCA). RESULTS In total, 235 patients diagnosed with CCRCC, comprising 168 low-grade and 67 high-grade tumors, were included in this study. A comparison of the predictive performances of different models revealed that the logistic regression model exhibited relatively good stability and robustness. The AUC of the combined model for the training, internal validation and external validation sets were 0.871, 0.785 and 0.826, respectively, which were higher than those of the clinical and imaging histology models. Furthermore, the calibration curve demonstrated excellent concordance between the predicted Fuhrman grade probability of CCRCC using the combined model and the observed values in both the training and validation sets. Additionally, within the threshold range of 0-0.93, the combined model demonstrated substantial clinical utility, as evidenced by DCA. CONCLUSION The application of US radiomics techniques enabled objective prediction of Fuhrman grading in patients with CCRCC. Nevertheless, certain clinical indicators remain indispensable, underscoring the pressing need for their integrated use in clinical practice. ADVANCES IN KNOWLEDGE Previous studies have predominantly focused on using computed tomography or magnetic resonance imaging modalities to predict the Fuhrman grade of CCRCC. Our findings demonstrate that a prediction model based on US images is more cost-effective, easily accessible and exhibits commendable performance. Consequently, this study offers a promising approach to maximizing the use of US examinations in future research.
Collapse
Affiliation(s)
- Youchang Yang
- Department of Radiology, Qingdao Medical and Industrial Cross Key Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Ziyi Yuan
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qingguo Ren
- Department of Radiology, Qingdao Medical and Industrial Cross Key Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Jiajia Wang
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuai Guan
- Department of Radiology, Qingdao Medical and Industrial Cross Key Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Xiaoqiang Tang
- Department of Radiology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Qingjun Jiang
- Department of Radiology, Qingdao Medical and Industrial Cross Key Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Xiangshui Meng
- Department of Radiology, Qingdao Medical and Industrial Cross Key Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China.
| |
Collapse
|
34
|
Cai W, Guo K, Chen Y, Shi Y, Chen J. Sub-regional CT Radiomics for the Prediction of Ki-67 Proliferation Index in Gastrointestinal Stromal Tumors: A Multi-center Study. Acad Radiol 2024; 31:4974-4984. [PMID: 39033048 DOI: 10.1016/j.acra.2024.06.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/18/2024] [Accepted: 06/22/2024] [Indexed: 07/23/2024]
Abstract
RATIONALE AND OBJECTIVES The objective was to assess and examine radiomics models derived from contrast-enhanced CT for their predictive capacity using the sub-regional radiomics regarding the Ki-67 proliferation index (PI) in patients with pathologically confirmed gastrointestinal stromal tumors (GIST). METHODS In this retrospective study, a total of 412 GIST patients across three institutions (223 from center 1, 106 from center 2, and 83 from center 3) was enrolled. Radiomic features were derived from various sub-regions of the tumor region of interest employing the K-means approach. The Least Absolute Shrinkage and Selection Operator (LASSO) regression was employed to identify features correlated with Ki-67 PI level in GIST patients. A support vector machine (SVM) model was then constructed to predict the high level of Ki-67 (Ki-67 index >8%), drawing on the radiomics features from each sub-region within the training cohort. RESULTS After features selection process, 6, 9, 9, 7 features were obtained to construct SVM models based on sub-region 1, 2, 3 and the entire tumor, respectively. Among different models, the model developed by the sub-region 1 achieved an area under the receiver operating characteristic curve (AUC) of 0.880 (95% confidence interval [CI]: 0.830 to 0.919), 0.852 (95% CI: 0.770-0.914), 0.799 (95% CI: 0.697-0.879) in the training, external test set 1, and 2, respectively. CONCLUSION The results of the present study suggested that SVM model based on the sub-regional radiomics features had the potential of predicting Ki-67 PI level in patients with GIST.
Collapse
Affiliation(s)
- Wemin Cai
- Department of Emergency, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou 325000, China; Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Kun Guo
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yongxian Chen
- Department of Chest cancer, Xiamen Second People's Hospital, Xiamen 36100, China
| | - Yubo Shi
- Department of Pulmonary, Yueqing People's Hospital, Wenzhou 325000, China
| | - Junkai Chen
- Department of Emergency, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou 325000, China.
| |
Collapse
|
35
|
Jiang L, Zhou Y, Miao W, Zhu H, Zou N, Tian Y, Pan H, Jin W, Huang J, Luo Q. Artificial intelligence-assisted quantitative CT parameters in predicting the degree of risk of solitary pulmonary nodules. Ann Med 2024; 56:2405075. [PMID: 39297299 DOI: 10.1080/07853890.2024.2405075] [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: 12/11/2023] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 09/21/2024] Open
Abstract
INTRODUCTION Artificial intelligence (AI) shows promise for evaluating solitary pulmonary nodules (SPNs) on computed tomography (CT). Accurately determining cancer invasiveness can guide treatment. We aimed to investigate quantitative CT parameters for invasiveness prediction. METHODS Patients with stage 0-IB NSCLC after surgical resection were retrospectively analysed. Preoperative CTs were evaluated with specialized software for nodule segmentation and CT quantification. Pathology was the reference for invasiveness. Univariate and multivariate logistic regression assessed predictors of high-risk SPN. RESULTS Three hundred and fifty-five SPN were included. On multivariate analysis, CT value mean and nodule type (ground glass opacity vs. solid) were independent predictors of high-risk SPN. The area under the curve (AUC) was 0.811 for identifying high-risk nodules. CONCLUSIONS Quantitative CT measures and nodule type correlated with invasiveness. Software-based CT assessment shows potential for noninvasive prediction to guide extent of resection. Further prospective validation is needed, including comparison with benign nodules.
Collapse
Affiliation(s)
- Long Jiang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Zhou
- Department of Purchasing Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wang Miao
- Department of Thoracic Surgery, The Third People's Hospital of Zhengzhou, Zhengzhou, China
| | - Hongda Zhu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ningyuan Zou
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Tian
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanbo Pan
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqiu Jin
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Huang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingquan Luo
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Tarquino J, Rodríguez J, Becerra D, Roa-Peña L, Romero E. Engineered feature embeddings meet deep learning: A novel strategy to improve bone marrow cell classification and model transparency. J Pathol Inform 2024; 15:100390. [PMID: 39712979 PMCID: PMC11662281 DOI: 10.1016/j.jpi.2024.100390] [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: 05/24/2024] [Revised: 06/18/2024] [Accepted: 06/29/2024] [Indexed: 12/24/2024] Open
Abstract
Cytomorphology evaluation of bone marrow cell is the initial step to diagnose different hematological diseases. This assessment is still manually performed by trained specialists, who may be a bottleneck within the clinical process. Deep learning algorithms are a promising approach to automate this bone marrow cell evaluation. These artificial intelligence models have focused on limited cell subtypes, mainly associated to a particular disease, and are frequently presented as black boxes. The herein introduced strategy presents an engineered feature representation, the region-attention embedding, which improves the deep learning classification performance of a cytomorphology with 21 bone marrow cell subtypes. This embedding is built upon a specific organization of cytology features within a squared matrix by distributing them after pre-segmented cell regions, i.e., cytoplasm, nucleus, and whole-cell. This novel cell image representation, aimed to preserve spatial/regional relations, is used as input of the network. Combination of region-attention embedding and deep learning networks (Xception and ResNet50) provides local relevance associated to image regions, adding up interpretable information to the prediction. Additionally, this approach is evaluated in a public database with the largest number of cell subtypes (21) by a thorough evaluation scheme with three iterations of a 3-fold cross-validation, performed in 80% of the images (n = 89,484), and a testing process in an unseen set of images composed by the remaining 20% of the images (n = 22,371). This evaluation process demonstrates the introduced strategy outperforms previously published approaches in an equivalent validation set, with a f1-score of 0.82, and presented competitive results on the unseen data partition with a f1-score of 0.56.
Collapse
Affiliation(s)
- Jonathan Tarquino
- Computer Imaging and Medical Application Laboratory, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| | - Jhonathan Rodríguez
- Computer Imaging and Medical Application Laboratory, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| | - David Becerra
- Department of Pathology, School of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Lucia Roa-Peña
- Department of Pathology, School of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Eduardo Romero
- Computer Imaging and Medical Application Laboratory, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| |
Collapse
|
37
|
Al Hasan MM, Ghazimoghadam S, Tunlayadechanont P, Mostafiz MT, Gupta M, Roy A, Peters K, Hochhegger B, Mancuso A, Asadizanjani N, Forghani R. Automated Segmentation of Lymph Nodes on Neck CT Scans Using Deep Learning. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:2955-2966. [PMID: 38937342 PMCID: PMC11612088 DOI: 10.1007/s10278-024-01114-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 06/29/2024]
Abstract
Early and accurate detection of cervical lymph nodes is essential for the optimal management and staging of patients with head and neck malignancies. Pilot studies have demonstrated the potential for radiomic and artificial intelligence (AI) approaches in increasing diagnostic accuracy for the detection and classification of lymph nodes, but implementation of many of these approaches in real-world clinical settings would necessitate an automated lymph node segmentation pipeline as a first step. In this study, we aim to develop a non-invasive deep learning (DL) algorithm for detecting and automatically segmenting cervical lymph nodes in 25,119 CT slices from 221 normal neck contrast-enhanced CT scans from patients without head and neck cancer. We focused on the most challenging task of segmentation of small lymph nodes, evaluated multiple architectures, and employed U-Net and our adapted spatial context network to detect and segment small lymph nodes measuring 5-10 mm. The developed algorithm achieved a Dice score of 0.8084, indicating its effectiveness in detecting and segmenting cervical lymph nodes despite their small size. A segmentation framework successful in this task could represent an essential initial block for future algorithms aiming to evaluate small objects such as lymph nodes in different body parts, including small lymph nodes looking normal to the naked human eye but harboring early nodal metastases.
Collapse
Affiliation(s)
- Md Mahfuz Al Hasan
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Electrical and Computer Engineering, University of Florida College of Medicine, Gainesville, FL, USA
| | - Saba Ghazimoghadam
- Augmented Intelligence and Precision Health Laboratory, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Padcha Tunlayadechanont
- Augmented Intelligence and Precision Health Laboratory, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Diagnostic and Therapeutic Radiology and Research, Faculty of Medicine Ramathibodi Hospital, Ratchathewi, Bangkok, Thailand
| | - Mohammed Tahsin Mostafiz
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Electrical and Computer Engineering, University of Florida College of Medicine, Gainesville, FL, USA
| | - Manas Gupta
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
| | - Antika Roy
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Electrical and Computer Engineering, University of Florida College of Medicine, Gainesville, FL, USA
| | - Keith Peters
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Bruno Hochhegger
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Anthony Mancuso
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Navid Asadizanjani
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Electrical and Computer Engineering, University of Florida College of Medicine, Gainesville, FL, USA
| | - Reza Forghani
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA.
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, USA.
- Division of Medical Physics, University of Florida College of Medicine, Gainesville, FL, USA.
- Department of Neurology, Division of Movement Disorders, University of Florida College of Medicine, Gainesville, FL, USA.
- Augmented Intelligence and Precision Health Laboratory, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
38
|
Lee H, Chung JW, Yun SC, Jung SW, Yoon YJ, Kim JH, Cha B, Kayasseh MA, Kim KO. Validation of Artificial Intelligence Computer-Aided Detection on Gastric Neoplasm in Upper Gastrointestinal Endoscopy. Diagnostics (Basel) 2024; 14:2706. [PMID: 39682614 DOI: 10.3390/diagnostics14232706] [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: 11/14/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Gastric cancer ranks fifth for incidence and fourth in the leading causes of mortality worldwide. In this study, we aimed to validate previously developed artificial intelligence (AI) computer-aided detection (CADe) algorithm, called ALPHAON® in detecting gastric neoplasm. METHODS We used the retrospective data of 500 still images, including 5 benign gastric ulcers, 95 with gastric cancer, and 400 normal images. Thereby we validated the CADe algorithm measuring accuracy, sensitivity, and specificity with the result of receiver operating characteristic curves (ROC) and area under curve (AUC) in addition to comparing the diagnostic performance status of four expert endoscopists, four trainees, and four beginners from two university-affiliated hospitals with CADe algorithm. After a washing-out period of over 2 weeks, endoscopists performed gastric detection on the same dataset of the 500 endoscopic images again marked by ALPHAON®. RESULTS The CADe algorithm presented high validity in detecting gastric neoplasm with accuracy (0.88, 95% CI: 0.85 to 0.91), sensitivity (0.93, 95% CI: 0.88 to 0.98), specificity (0.87, 95% CI: 0.84 to 0.90), and AUC (0.962). After a washing-out period of over 2 weeks, overall validity improved in the trainee and beginner groups with the assistance of ALPHAON®. Significant improvement was present, especially in the beginner group (accuracy 0.94 (0.93 to 0.96) p < 0.001, sensitivity 0.87 (0.82 to 0.92) p < 0.001, specificity 0.96 (0.95 to 0.97) p < 0.001). CONCLUSIONS The high validation performance state of the CADe algorithm system was verified. Also, ALPHAON® has demonstrated its potential to serve as an endoscopic educator for beginners improving and making progress in sensitivity and specificity.
Collapse
Affiliation(s)
- Hannah Lee
- Division of Gastroenterology, Department of Internal Medicine, Gachon University, Gil Medical Center, Incheon 21565, Republic of Korea
| | - Jun-Won Chung
- Division of Gastroenterology, Department of Internal Medicine, Gachon University, Gil Medical Center, Incheon 21565, Republic of Korea
| | - Sung-Cheol Yun
- Division of Biostatistics, Center for Medical Research and Information, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Sung Woo Jung
- Division of Gastroenterology, Department of Internal Medicine, Korea University College of Medicine, Ansan 15355, Republic of Korea
| | | | - Ji Hee Kim
- CAIMI Co., Ltd., Incheon 22004, Republic of Korea
| | - Boram Cha
- Division of Gastroenterology, Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon 22332, Republic of Korea
| | - Mohd Azzam Kayasseh
- Division of Gastroenterology, Dr. Sulaiman AI Habib Medical Group, Dubai Healthcare City, Dubai 51431, United Arab Emirates
| | - Kyoung Oh Kim
- Division of Gastroenterology, Department of Internal Medicine, Gachon University, Gil Medical Center, Incheon 21565, Republic of Korea
| |
Collapse
|
39
|
Jiang X, Hu J, Jiang Q, Zhou T, Yao F, Sun Y, Zhou C, Ma Q, Zhao J, Shi K, Yang W, Zhou X, Wang Y, Liu S, Xin X, Fan L. CT-based whole lung radiomics nomogram to identify middle-aged and elderly COVID-19 patients at high risk of progressing to critical disease. J Appl Clin Med Phys 2024:e14562. [PMID: 39611805 DOI: 10.1002/acm2.14562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 08/19/2024] [Accepted: 09/16/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND COVID-19 remains widespread and poses a threat to people's physical and mental health, especially middle-aged and elderly individuals. Early identification of COVID-19 patients at high risk of progressing to critical disease helps improve overall patient outcomes and healthcare efficiency. PURPOSE To develop a radiomics nomogram to predict the risk of newly admitted middle-aged and elderly COVID-19 patients progressing to critical disease. METHODS A total of 794 patients (aged 40 years or above) were retrospectively included in the study from two institutions, all of them were with non-critical COVID-19 on admission. At follow-up, patients were divided into non-critical group and critical group. About 443 patients (384 non-critical and 59 critical) from the first hospital were randomly assigned to the training (n = 311) and internal validation (n = 132) set in a 7:3 ratio. Additionally, an independent external cohort of 351 patients (292 non-critical and 59 critical) from another hospital was evaluated. Radiomics signatures and clinical indicators were used to build a radiomics model and a clinical model after computed tomography (CT) image processing, CT whole-lung segmentation, feature extraction, and feature selection. The radiomics nomogram model integrated radiomics model and clinical model. The receiver operating characteristic curve (AUC) was used to assess the performance of the proposed models. Calibration curves and decision curve analysis were used to assess the performance of the radiomics nomogram. RESULTS For the training, internal validation, and external validation sets, the AUC values of the radiomic nomogram for the prediction of COVID-19 progression were 0.916, 0.917, and 0.890, respectively. Calibration curves indicated that there was no significant departure between prediction and observation in three sets. The decision curve image demonstrated the clinical utility of the nomogram model. CONCLUSIONS Our nomogram model incorporates radiomics features and clinical indicators, it provides a new pathway to increase predictive accuracy or clinical utility, further helping to provide personalized management for middle-aged and elderly patients with COVID-19.
Collapse
Affiliation(s)
- Xin'ang Jiang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jun Hu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Qinling Jiang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Taohu Zhou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
- School of Medical Imaging, Weifang Medical University, Weifang, Shandong, China
| | - Fei Yao
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Yi Sun
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Chao Zhou
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Qianyun Ma
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jingyi Zhao
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Kang Shi
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wen Yang
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xiuxiu Zhou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yun Wang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shiyuan Liu
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiaoyan Xin
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Li Fan
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
40
|
Bachnas MA, Andonotopo W, Dewantiningrum J, Adi Pramono MB, Stanojevic M, Kurjak A. The utilization of artificial intelligence in enhancing 3D/4D ultrasound analysis of fetal facial profiles. J Perinat Med 2024; 52:899-913. [PMID: 39383043 DOI: 10.1515/jpm-2024-0347] [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: 08/02/2024] [Accepted: 09/05/2024] [Indexed: 10/11/2024]
Abstract
Artificial intelligence (AI) has emerged as a transformative technology in the field of healthcare, offering significant advancements in various medical disciplines, including obstetrics. The integration of artificial intelligence into 3D/4D ultrasound analysis of fetal facial profiles presents numerous benefits. By leveraging machine learning and deep learning algorithms, AI can assist in the accurate and efficient interpretation of complex 3D/4D ultrasound data, enabling healthcare providers to make more informed decisions and deliver better prenatal care. One such innovation that has significantly improved the analysis of fetal facial profiles is the integration of AI in 3D/4D ultrasound imaging. In conclusion, the integration of artificial intelligence in the analysis of 3D/4D ultrasound data for fetal facial profiles offers numerous benefits, including improved accuracy, consistency, and efficiency in prenatal diagnosis and care.
Collapse
Affiliation(s)
- Muhammad Adrianes Bachnas
- Fetomaternal Division, Department of Obstetrics and Gynecology, Medical Faculty of Sebelas Maret University, Moewardi Hospital, Solo, Surakarta, Indonesia
| | - Wiku Andonotopo
- Fetomaternal Division, Department of Obstetrics and Gynecology, Ekahospital BSD City, Serpong, Tangerang, Banten, Indonesia
| | - Julian Dewantiningrum
- Fetomaternal Division, Department of Obstetrics and Gynecology, Medical Faculty of Diponegoro University, Dr. Kariadi Hospital, Semarang, Indonesia
| | - Mochammad Besari Adi Pramono
- Fetomaternal Division, Department of Obstetrics and Gynecology, Medical Faculty of Diponegoro University, Dr. Kariadi Hospital, Semarang, Indonesia
| | - Milan Stanojevic
- Department of Obstetrics and Gynecology, Medical School University of Zagreb, Zagreb, Croatia
| | - Asim Kurjak
- Department of Obstetrics and Gynecology, Medical School University of Zagreb, Zagreb, Croatia
| |
Collapse
|
41
|
Bartl-Pokorny KD, Zitta C, Beirit M, Vogrinec G, Schuller BW, Pokorny FB. Focused review on artificial intelligence for disease detection in infants. Front Digit Health 2024; 6:1459640. [PMID: 39654981 PMCID: PMC11625793 DOI: 10.3389/fdgth.2024.1459640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/30/2024] [Indexed: 12/12/2024] Open
Abstract
Over the last years, studies using artificial intelligence (AI) for the detection and prediction of diseases have increased and also concentrated more and more on vulnerable groups of individuals, such as infants. The release of ChatGPT demonstrated the potential of large language models (LLMs) and heralded a new era of AI with manifold application possibilities. However, the impact of this new technology on medical research cannot be fully estimated yet. In this work, we therefore aimed to summarise the most recent pre-ChatGPT developments in the field of automated detection and prediction of diseases and disease status in infants, i.e., within the first 12 months of life. For this, we systematically searched the scientific databases PubMed and IEEE Xplore for original articles published within the last five years preceding the release of ChatGPT (2018-2022). The search revealed 927 articles; a final number of 154 articles was included for review. First of all, we examined research activity over time. Then, we analysed the articles from 2022 for medical conditions, data types, tasks, AI approaches, and reported model performance. A clear trend of increasing research activity over time could be observed. The most recently published articles focused on medical conditions of twelve different ICD-11 categories; "certain conditions originating in the perinatal period" was the most frequently addressed disease category. AI models were trained with a variety of data types, among which clinical and demographic information and laboratory data were most frequently exploited. The most frequently performed tasks aimed to detect present diseases, followed by the prediction of diseases and disease status at a later point in development. Deep neural networks turned out as the most popular AI approach, even though traditional methods, such as random forests and support vector machines, still play a role-presumably due to their explainability or better suitability when the amount of data is limited. Finally, the reported performances in many of the reviewed articles suggest that AI has the potential to assist in diagnostic procedures for infants in the near future. LLMs will boost developments in this field in the upcoming years.
Collapse
Affiliation(s)
- Katrin D. Bartl-Pokorny
- Division of Phoniatrics, Medical University of Graz, Graz, Austria
- EIHW – Chair of Embedded Intelligence for Health Care and Wellbeing, University of Augsburg, Augsburg, Germany
- CHI – Chair of Health Informatics, Technical University of Munich, Munich, Germany
| | - Claudia Zitta
- Division of Phoniatrics, Medical University of Graz, Graz, Austria
| | - Markus Beirit
- EIHW – Chair of Embedded Intelligence for Health Care and Wellbeing, University of Augsburg, Augsburg, Germany
| | - Gunter Vogrinec
- Division of Phoniatrics, Medical University of Graz, Graz, Austria
| | - Björn W. Schuller
- EIHW – Chair of Embedded Intelligence for Health Care and Wellbeing, University of Augsburg, Augsburg, Germany
- CHI – Chair of Health Informatics, Technical University of Munich, Munich, Germany
- Center for Interdisciplinary Health Research, University of Augsburg, Augsburg, Germany
- Munich Center for Machine Learning (MCML), Munich, Germany
- GLAM – Group on Language, Audio & Music, Imperial College London, London, United Kingdom
| | - Florian B. Pokorny
- Division of Phoniatrics, Medical University of Graz, Graz, Austria
- EIHW – Chair of Embedded Intelligence for Health Care and Wellbeing, University of Augsburg, Augsburg, Germany
- CHI – Chair of Health Informatics, Technical University of Munich, Munich, Germany
- Center for Interdisciplinary Health Research, University of Augsburg, Augsburg, Germany
- Munich Center for Machine Learning (MCML), Munich, Germany
| |
Collapse
|
42
|
Crimì F, D’Alessandro C, Zanon C, Celotto F, Salvatore C, Interlenghi M, Castiglioni I, Quaia E, Pucciarelli S, Spolverato G. A Machine Learning Model Based on MRI Radiomics to Predict Response to Chemoradiation Among Patients with Rectal Cancer. Life (Basel) 2024; 14:1530. [PMID: 39768239 PMCID: PMC11677041 DOI: 10.3390/life14121530] [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: 09/29/2024] [Revised: 11/16/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND With rectum-sparing protocols becoming more common for rectal cancer treatment, this study aimed to predict the pathological complete response (pCR) to preoperative chemoradiotherapy (pCRT) in rectal cancer patients using pre-treatment MRI and a radiomics-based machine learning approach. METHODS We divided MRI-data from 102 patients into a training cohort (n = 72) and a validation cohort (n = 30). In the training cohort, 52 patients were classified as non-responders and 20 as pCR based on histological results from total mesorectal excision. RESULTS We trained various machine learning models using radiomic features to capture disease heterogeneity between responders and non-responders. The best-performing model achieved a receiver operating characteristic area under the curve (ROC-AUC) of 73% and an accuracy of 70%, with a sensitivity of 78% and a positive predictive value (PPV) of 80%. In the validation cohort, the model showed a sensitivity of 81%, specificity of 75%, and accuracy of 80%. CONCLUSIONS These results highlight the potential of radiomics and machine learning in predicting treatment response and support the integration of advanced imaging and computational methods for personalized rectal cancer management.
Collapse
Affiliation(s)
- Filippo Crimì
- Institute of Radiology, Department of Medicine-DIMED, University of Padova, 35128 Padova, Italy; (F.C.); (C.D.); (C.Z.); (E.Q.)
| | - Carlo D’Alessandro
- Institute of Radiology, Department of Medicine-DIMED, University of Padova, 35128 Padova, Italy; (F.C.); (C.D.); (C.Z.); (E.Q.)
| | - Chiara Zanon
- Institute of Radiology, Department of Medicine-DIMED, University of Padova, 35128 Padova, Italy; (F.C.); (C.D.); (C.Z.); (E.Q.)
| | - Francesco Celotto
- Third Surgical Clinic, Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG), University of Padova, 35128 Padova, Italy; (F.C.); (S.P.)
| | - Christian Salvatore
- DeepTrace Technologies S.R.L., Via Conservatorio 17, 20122 Milano, Italy; (C.S.); (M.I.)
- Department of Science, Technology and Society, University School for Advanced Studies IUSS Pavia, 27100 Pavia, Italy
| | - Matteo Interlenghi
- DeepTrace Technologies S.R.L., Via Conservatorio 17, 20122 Milano, Italy; (C.S.); (M.I.)
| | - Isabella Castiglioni
- Dipartimento di Fisica Giuseppe Occhialini, Università degli Studi di Milano Bicocca, Piazza della Scienza 3, 20126 Milano, Italy;
| | - Emilio Quaia
- Institute of Radiology, Department of Medicine-DIMED, University of Padova, 35128 Padova, Italy; (F.C.); (C.D.); (C.Z.); (E.Q.)
| | - Salvatore Pucciarelli
- Third Surgical Clinic, Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG), University of Padova, 35128 Padova, Italy; (F.C.); (S.P.)
| | - Gaya Spolverato
- Third Surgical Clinic, Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG), University of Padova, 35128 Padova, Italy; (F.C.); (S.P.)
| |
Collapse
|
43
|
Moro F, Ciancia M, Zace D, Vagni M, Tran HE, Giudice MT, Zoccoli SG, Mascilini F, Ciccarone F, Boldrini L, D'Antonio F, Scambia G, Testa AC. Role of artificial intelligence applied to ultrasound in gynecology oncology: A systematic review. Int J Cancer 2024; 155:1832-1845. [PMID: 38989809 DOI: 10.1002/ijc.35092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/03/2024] [Accepted: 06/27/2024] [Indexed: 07/12/2024]
Abstract
The aim of this paper was to explore the role of artificial intelligence (AI) applied to ultrasound imaging in gynecology oncology. Web of Science, PubMed, and Scopus databases were searched. All studies were imported to RAYYAN QCRI software. The overall quality of the included studies was assessed using QUADAS-AI tool. Fifty studies were included, of these 37/50 (74.0%) on ovarian masses or ovarian cancer, 5/50 (10.0%) on endometrial cancer, 5/50 (10.0%) on cervical cancer, and 3/50 (6.0%) on other malignancies. Most studies were at high risk of bias for subject selection (i.e., sample size, source, or scanner model were not specified; data were not derived from open-source datasets; imaging preprocessing was not performed) and index test (AI models was not externally validated) and at low risk of bias for reference standard (i.e., the reference standard correctly classified the target condition) and workflow (i.e., the time between index test and reference standard was reasonable). Most studies presented machine learning models (33/50, 66.0%) for the diagnosis and histopathological correlation of ovarian masses, while others focused on automatic segmentation, reproducibility of radiomics features, improvement of image quality, prediction of therapy resistance, progression-free survival, and genetic mutation. The current evidence supports the role of AI as a complementary clinical and research tool in diagnosis, patient stratification, and prediction of histopathological correlation in gynecological malignancies. For example, the high performance of AI models to discriminate between benign and malignant ovarian masses or to predict their specific histology can improve the diagnostic accuracy of imaging methods.
Collapse
Affiliation(s)
- Francesca Moro
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Marianna Ciancia
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Salute della Donna e del Bambino, Università degli studi di Padova, Padova, Italy
| | - Drieda Zace
- Infectious Disease Clinic, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Marica Vagni
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Huong Elena Tran
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Maria Teresa Giudice
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Sofia Gambigliani Zoccoli
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Floriana Mascilini
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Francesca Ciccarone
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Luca Boldrini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | | | - Giovanni Scambia
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonia Carla Testa
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
44
|
Ma T, Wang J, Ma F, Shi J, Li Z, Cui J, Wu G, Zhao G, An Q. Visualization analysis of research hotspots and trends in MRI-based artificial intelligence in rectal cancer. Heliyon 2024; 10:e38927. [PMID: 39524896 PMCID: PMC11544045 DOI: 10.1016/j.heliyon.2024.e38927] [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: 03/02/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
Background Rectal cancer (RC) is one of the most common types of cancer worldwide. With the development of artificial intelligence (AI), the application of AI in preoperative evaluation and follow-up treatment of RC based on magnetic resonance imaging (MRI) has been the focus of research in this field. This review was conducted to develop comprehensive insight into the current research progress, hotspots, and future trends in AI based on MRI in RC, which remains to be studied. Methods Literature related to AI based on MRI and RC, as of November 2023, was obtained from the Web of Science Core Collection database. Visualization and bibliometric analyses of publication quantity and content were conducted to explore temporal trends, spatial distribution, collaborative networks, influential articles, keyword co-occurrence, and research directions. Results A total of 177 papers (152 original articles and 25 reviews) were identified from 24 countries/regions, 351 institutions, and 81 journals. Since 2019, the number of studies on this topic has rapidly increased. China and the United States have contributed the highest number of publications and institutions, cultivating the most intimate collaborative relationship. The highest number of articles derive from Sun Yat-sen University, while Frontiers in Oncology has published the highest number of relevant articles. Research on MRI-based AI in this field has mainly focused on preoperative diagnosis and prediction of treatment efficacy and prognosis. Conclusions This study provides an objective and comprehensive overview of the publications on MRI-based AI in RC and identifies the present research landscape, hotspots, and prospective trends in this field, which can provide valuable guidance for scholars worldwide.
Collapse
Affiliation(s)
- Tianming Ma
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jiawen Wang
- Department of Urology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Fuhai Ma
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jinxin Shi
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Zijian Li
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jian Cui
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Guoju Wu
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Gang Zhao
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Qi An
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| |
Collapse
|
45
|
Dai T, Gu QB, Peng YJ, Yu CL, Liu P, He YQ. Preoperative Noninvasive Prediction of Recurrence-Free Survival in Hepatocellular Carcinoma Using CT-Based Radiomics Model. J Hepatocell Carcinoma 2024; 11:2211-2222. [PMID: 39558966 PMCID: PMC11571988 DOI: 10.2147/jhc.s493044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/09/2024] [Indexed: 11/20/2024] Open
Abstract
Purpose This study aims to explore the value of radiomics combined with clinical parameters in predicting recurrence-free survival (RFS) after the resection of hepatocellular carcinoma (HCC). Patients and Methods In this retrospective study, a total of 322 patients with HCC who underwent contrast-enhanced computed tomography (CT) and radical surgical resection were enrolled and randomly divided into a training group (n = 223) and a validation group (n = 97). In the training group, Univariate and multivariate Cox regression analyses were employed to obtain clinical variables related to RFS for constructing the clinical model. The least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression analyses were employed to construct the radiomics model, and the clinical-radiomics model was further constructed. Model prediction performance was subsequently assessed by the area under the time-dependent receiver operating characteristic curve (AUC) and calibration curve. Additionally, Kaplan-Meier analysis was used to evaluate the model's value in predicting RFS. Correlations between radiomics features and pathological parameters were analyzed. Results The clinical-radiomics model predicted RFS at 1, 2, and 3 years more accurately than the clinical or radiomics model alone (training group, AUC = 0.834, 0.765 and 0.831, respectively; validation group, AUC = 0.715, 0.710 and 0.793, respectively). The predicted high-risk subgroup based on the clinical-radiomics nomogram had shorter RFS than predicted low-risk subgroup in data sets, enabling risk stratification of various clinical subgroups. Correlation analysis revealed that the rad-score was positively related to microvascular invasion (MVI) and Edmondson-Steiner grade. Conclusion The clinical-radiomics model effectively predicts RFS in HCC patients and identifies high-risk individuals for recurrence.
Collapse
Affiliation(s)
- Ting Dai
- Department of Radiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, People’s Republic of China
| | - Qian-Biao Gu
- Department of Radiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, People’s Republic of China
| | - Ying-Jie Peng
- Department of Radiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, People’s Republic of China
| | - Chuan-Lin Yu
- Department of Radiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, People’s Republic of China
| | - Peng Liu
- Department of Radiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, People’s Republic of China
| | - Ya-Qiong He
- Department of Radiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, People’s Republic of China
| |
Collapse
|
46
|
Sun X, Li S, Ma C, Fang W, Jing X, Yang C, Li H, Zhang X, Ge C, Liu B, Li Z. Glioma subtype prediction based on radiomics of tumor and peritumoral edema under automatic segmentation. Sci Rep 2024; 14:27471. [PMID: 39523433 PMCID: PMC11551193 DOI: 10.1038/s41598-024-79344-9] [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: 07/08/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
Comprehensive and non-invasive preoperative molecular diagnosis is important for prognostic and therapy decision-making in adult-type diffuse gliomas. We employed a deep learning method for automatic segmentation of brain gliomas directly from conventional magnetic resonance imaging (MRI) scans of the tumor core and peritumoral edema regions based on available glioma MRI data provided in the BraTS2021. Three-dimensional volumes of interest were segmented from 424 cases of glioma imaging data retrospectively obtained from two medical centers using the segmentation method and radiomic features were extracted. We developed a subtype prediction model based on extracted radiomic features and analyzed significance and correlations between glioma morphological characteristics and pathological features using data from patients with adult-type diffuse glioma. The automated segmentation achieved mean Dice scores of 0.884 and 0.889 for the tumor core and whole tumor, respectively. The area under the receiver operating characteristic curve for the prediction of adult-type diffuse gliomas subtypes was 0.945. "Glioblastoma, IDH-wildtype", "Astrocytoma, IDH-mutant", and "Oligodendroglioma, IDH-mutant, 1p/19q-coded" showed AUCs of 0.96, 0.914, and 0.961, respectively, for subtype prediction. Glioma morphological characteristics, molecular and pathological levels, and clinical data showed significant differences and correlations. An automatic segmentation model for gliomas based on 3D U-Nets was developed, and the prediction model for gliomas built using the parameters obtained from the automatic segmentation model showed high overall performance.
Collapse
Affiliation(s)
- Xiangyu Sun
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.125 Donghu Road, WuChang, Wuhan, 430062, China
| | - Sirui Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuchang District, Wuhan, China
| | - Chao Ma
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.125 Donghu Road, WuChang, Wuhan, 430062, China
| | - Wei Fang
- Wuhan Zhongke Industrial Research Institute of Medical Science Co., Ltd., Wuhan, China
| | - Xin Jing
- Wuhan United Imaging Healthcare Surgical Technology Co., Ltd., Wuhan, China
| | - Chao Yang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.125 Donghu Road, WuChang, Wuhan, 430062, China
| | - Huan Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuchang District, Wuhan, China
| | - Xu Zhang
- Wuhan United Imaging Healthcare Surgical Technology Co., Ltd., Wuhan, China
| | - Chuanbin Ge
- Wuhan United Imaging Healthcare Surgical Technology Co., Ltd., Wuhan, China
| | - Bo Liu
- Wuhan United Imaging Healthcare Surgical Technology Co., Ltd., Wuhan, China
| | - Zhiqiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.125 Donghu Road, WuChang, Wuhan, 430062, China.
| |
Collapse
|
47
|
Jacob M, Reddy RP, Garcia RI, Reddy AP, Khemka S, Roghani AK, Pattoor V, Sehar U, Reddy PH. Harnessing Artificial Intelligence for the Detection and Management of Colorectal Cancer Treatment. Cancer Prev Res (Phila) 2024; 17:499-515. [PMID: 39077801 PMCID: PMC11534518 DOI: 10.1158/1940-6207.capr-24-0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/26/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
Currently, eight million people in the United States suffer from cancer and it is a major global health concern. Early detection and interventions are urgently needed for all cancers, including colorectal cancer. Colorectal cancer is the third most common type of cancer worldwide. Based on the diagnostic efforts to general awareness and lifestyle choices, it is understandable why colorectal cancer is so prevalent today. There is a notable lack of awareness concerning the impact of this cancer and its connection to lifestyle elements, as well as people sometimes mistaking symptoms for a different gastrointestinal condition. Artificial intelligence (AI) may assist in the early detection of all cancers, including colorectal cancer. The usage of AI has exponentially grown in healthcare through extensive research, and since clinical implementation, it has succeeded in improving patient lifestyles, modernizing diagnostic processes, and innovating current treatment strategies. Numerous challenges arise for patients with colorectal cancer and oncologists alike during treatment. For initial screening phases, conventional methods often result in misdiagnosis. Moreover, after detection, determining the course of which colorectal cancer can sometimes contribute to treatment delays. This article touches on recent advancements in AI and its clinical application while shedding light on why this disease is so common today.
Collapse
Affiliation(s)
- Michael Jacob
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas
| | - Ruhananhad P Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- Lubbock High School, Lubbock, Texas
| | - Ricardo I Garcia
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Aananya P Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- Lubbock High School, Lubbock, Texas
| | - Sachi Khemka
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Aryan Kia Roghani
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- Frenship High School, Lubbock, Texas
| | - Vasanthkumar Pattoor
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- University of South Florida, Tampa, Florida
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- Nutritional Sciences Department, College of Human Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, Texas
- Department of Speech, Language and Hearing Services, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, Texas
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas
| |
Collapse
|
48
|
Shenouda M, Shaikh A, Deutsch I, Mitchell O, Kindler HL, Armato SG. Radiomics for differentiation of somatic BAP1 mutation on CT scans of patients with pleural mesothelioma. J Med Imaging (Bellingham) 2024; 11:064501. [PMID: 39669009 PMCID: PMC11633667 DOI: 10.1117/1.jmi.11.6.064501] [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: 07/06/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024] Open
Abstract
Purpose The BRCA1-associated protein 1 (BAP1) gene is of great interest because somatic (BAP1) mutations are the most common alteration associated with pleural mesothelioma (PM). Further, germline mutation of the BAP1 gene has been linked to the development of PM. This study aimed to explore the potential of radiomics on computed tomography scans to identify somatic BAP1 gene mutations and assess the feasibility of radiomics in future research in identifying germline mutations. Approach A cohort of 149 patients with PM and known somatic BAP1 mutation status was collected, and a previously published deep learning model was used to first automatically segment the tumor, followed by radiologist modifications. Image preprocessing was performed, and texture features were extracted from the segmented tumor regions. The top features were selected and used to train 18 separate machine learning models using leave-one-out cross-validation (LOOCV). The performance of the models in distinguishing between BAP1-mutated (BAP1+) and BAP1 wild-type (BAP1-) tumors was evaluated using the receiver operating characteristic area under the curve (ROC AUC). Results A decision tree classifier achieved the highest overall AUC value of 0.69 (95% confidence interval: 0.60 and 0.77). The features selected most frequently through the LOOCV were all second-order (gray-level co-occurrence or gray-level size zone matrices) and were extracted from images with an applied transformation. Conclusions This proof-of-concept work demonstrated the potential of radiomics to differentiate among BAP1+/- in patients with PM. Future work will extend these methods to the assessment of germline BAP1 mutation status through image analysis for improved patient prognostication.
Collapse
Affiliation(s)
- Mena Shenouda
- The University of Chicago, Department of Radiology, Chicago, Illinois, United States
| | | | - Ilana Deutsch
- Northwestern University, Evanston, Illinois, United States
| | - Owen Mitchell
- The University of Chicago, Department of Medicine, Chicago, Illinois, United States
| | - Hedy L. Kindler
- The University of Chicago, Department of Medicine, Chicago, Illinois, United States
| | - Samuel G. Armato
- The University of Chicago, Department of Radiology, Chicago, Illinois, United States
| |
Collapse
|
49
|
Ueda D, Walston SL, Fujita S, Fushimi Y, Tsuboyama T, Kamagata K, Yamada A, Yanagawa M, Ito R, Fujima N, Kawamura M, Nakaura T, Matsui Y, Tatsugami F, Fujioka T, Nozaki T, Hirata K, Naganawa S. Climate change and artificial intelligence in healthcare: Review and recommendations towards a sustainable future. Diagn Interv Imaging 2024; 105:453-459. [PMID: 38918123 DOI: 10.1016/j.diii.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/27/2024]
Abstract
The rapid advancement of artificial intelligence (AI) in healthcare has revolutionized the industry, offering significant improvements in diagnostic accuracy, efficiency, and patient outcomes. However, the increasing adoption of AI systems also raises concerns about their environmental impact, particularly in the context of climate change. This review explores the intersection of climate change and AI in healthcare, examining the challenges posed by the energy consumption and carbon footprint of AI systems, as well as the potential solutions to mitigate their environmental impact. The review highlights the energy-intensive nature of AI model training and deployment, the contribution of data centers to greenhouse gas emissions, and the generation of electronic waste. To address these challenges, the development of energy-efficient AI models, the adoption of green computing practices, and the integration of renewable energy sources are discussed as potential solutions. The review also emphasizes the role of AI in optimizing healthcare workflows, reducing resource waste, and facilitating sustainable practices such as telemedicine. Furthermore, the importance of policy and governance frameworks, global initiatives, and collaborative efforts in promoting sustainable AI practices in healthcare is explored. The review concludes by outlining best practices for sustainable AI deployment, including eco-design, lifecycle assessment, responsible data management, and continuous monitoring and improvement. As the healthcare industry continues to embrace AI technologies, prioritizing sustainability and environmental responsibility is crucial to ensure that the benefits of AI are realized while actively contributing to the preservation of our planet.
Collapse
Affiliation(s)
- Daiju Ueda
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, Abeno-ku, Osaka 545-8585, Japan; Department of Artificial Intelligence, Graduate School of Medicine, Osaka Metropolitan University, Abeno-ku, Osaka 545-8585, Japan.
| | - Shannon L Walston
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, Abeno-ku, Osaka 545-8585, Japan
| | - Shohei Fujita
- Department of Radiology, University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yasutaka Fushimi
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, Sakyoku, Kyoto 606-8507, Japan
| | - Takahiro Tsuboyama
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Koji Kamagata
- Department of Radiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Akira Yamada
- Medical Data Science Course, Shinshu University School of Medicine, Matsumoto, Nagano 390-8621, Japan
| | - Masahiro Yanagawa
- Department of Radiology, Graduate School of Medicine, Osaka University, Suita-city, Osaka 565-0871, Japan
| | - Rintaro Ito
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Noriyuki Fujima
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Hokkaido 060-8648, Japan
| | - Mariko Kawamura
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Takeshi Nakaura
- Department of Diagnostic Radiology, Kumamoto University Graduate School of Medicine, Chuo-ku, Kumamoto 860-8556, Japan
| | - Yusuke Matsui
- Department of Radiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama 700-8558, Japan
| | - Fuminari Tatsugami
- Department of Diagnostic Radiology, Hiroshima University, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan
| | - Tomoyuki Fujioka
- Department of Diagnostic Radiology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Taiki Nozaki
- Department of Radiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kenji Hirata
- Department of Diagnostic Imaging, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Shinji Naganawa
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| |
Collapse
|
50
|
Zhang C, Zhang H, Zhang Q, Fan H, Yu P, Xia W, Zhang JZH, Liang X, Chen Y. Targeting ATP catalytic activity of chromodomain helicase CHD1L for the anticancer inhibitor discovery. Int J Biol Macromol 2024; 281:136678. [PMID: 39426766 DOI: 10.1016/j.ijbiomac.2024.136678] [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: 03/19/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
CHD1L functions as an ATP-dependent chromatin remodeling enzyme, featuring an ATPase catalytic domain activated by double-stranded DNA. Its involvement in critical aspects of cancer progression, such as drug resistance and epithelial-mesenchymal transition, underscores its potential as a promising therapeutic target for cancer treatment. In this study, we have pioneered an innovative approach that integrates multiple deep learning methodologies alongside biochemical and cellular experiments to identify promising inhibitors of CHD1L. Through virtual screening of over 1.5 million small molecule compounds, we carefully curated a set of 36 candidate compounds and rigorously evaluated the top 13 candidates. Our findings establish the lead compound C071-0684 as a potent anticancer agent with a novel molecular backbone, demonstrating remarkable efficacy against colorectal and breast cancer cells targeting CHD1L. This compound exhibited a comparable effect on ATPase activity and binding affinity with CHD1Li 6.11, highlighting its superior pharmacological potential. These results provide valuable insights and pave the way for the discovery and development of CHD1L-targeted therapeutics, holding great promise for cancer patients.
Collapse
Affiliation(s)
- Caiying Zhang
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Haiping Zhang
- Faculty of Synthetic Biology and Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qiuyun Zhang
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Hongjie Fan
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Pengfei Yu
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Wei Xia
- Faculty of Synthetic Biology and Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - John Z H Zhang
- Faculty of Synthetic Biology and Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xinmiao Liang
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China; CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Yang Chen
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China; CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| |
Collapse
|