1
|
Lu Z, Wang Y. Teaching CORnet human fMRI representations for enhanced model-brain alignment. Cogn Neurodyn 2025; 19:61. [PMID: 40242427 PMCID: PMC11999921 DOI: 10.1007/s11571-025-10252-y] [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: 10/23/2024] [Revised: 03/24/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Deep convolutional neural networks (DCNNs) have demonstrated excellent performance in object recognition and have been found to share some similarities with brain visual processing. However, the substantial gap between DCNNs and human visual perception still exists. Functional magnetic resonance imaging (fMRI) as a widely used technique in cognitive neuroscience can record neural activation in the human visual cortex during the process of visual perception. Can we teach DCNNs human fMRI signals to achieve a more brain-like model? To answer this question, this study proposed ReAlnet-fMRI, a model based on the SOTA vision model CORnet but optimized using human fMRI data through a multi-layer encoding-based alignment framework. This framework has been shown to effectively enable the model to learn human brain representations. The fMRI-optimized ReAlnet-fMRI exhibited higher similarity to the human brain than both CORnet and the control model in within- and across-subject as well as within- and across-modality model-brain (fMRI and EEG) alignment evaluations. Additionally, we conducted an in-depth analysis to investigate how the internal representations of ReAlnet-fMRI differ from CORnet in encoding various object dimensions. These findings provide the possibility of enhancing the brain-likeness of visual models by integrating human neural data, helping to bridge the gap between computer vision and visual neuroscience. Supplementary Information The online version contains supplementary material available at 10.1007/s11571-025-10252-y.
Collapse
Affiliation(s)
- Zitong Lu
- Departmen of Psychology, The Ohio State University, Columbus, 43210 USA
| | - Yile Wang
- Department of Neuroscience, The University of Texas at Dallas, Richardson, USA
| |
Collapse
|
2
|
Lei C, Sun W, Wang K, Weng R, Kan X, Li R. Artificial intelligence-assisted diagnosis of early gastric cancer: present practice and future prospects. Ann Med 2025; 57:2461679. [PMID: 39928093 PMCID: PMC11812113 DOI: 10.1080/07853890.2025.2461679] [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: 10/08/2024] [Revised: 12/09/2024] [Accepted: 01/23/2025] [Indexed: 02/11/2025] Open
Abstract
Gastric cancer (GC) occupies the first few places in the world among tumors in terms of incidence and mortality, causing serious harm to human health, and at the same time, its treatment greatly consumes the health care resources of all countries in the world. The diagnosis of GC is usually based on histopathologic examination, and it is very important to be able to detect and identify cancerous lesions at an early stage, but some endoscopists' lack of diagnostic experience and fatigue at work lead to a certain rate of under diagnosis. The rapid and striking development of Artificial intelligence (AI) has helped to enhance the ability to extract abnormal information from endoscopic images to some extent, and more and more researchers are applying AI technology to the diagnosis of GC. This initiative has not only improved the detection rate of early gastric cancer (EGC), but also significantly improved the survival rate of patients after treatment. This article reviews the results of various AI-assisted diagnoses of EGC in recent years, including the identification of EGC, the determination of differentiation type and invasion depth, and the identification of borders. Although AI has a better application prospect in the early diagnosis of ECG, there are still major challenges, and the prospects and limitations of AI application need to be further discussed.
Collapse
Affiliation(s)
- Changda Lei
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Wenqiang Sun
- Suzhou Medical College, Soochow University, Suzhou, China
- Department of Neonatology, Children’s Hospital of Soochow University, Suzhou, China
| | - Kun Wang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Ruixia Weng
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Xiuji Kan
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Rui Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
3
|
Peng D, Sun L, Zhou Q, Zhang Y. AI-driven approaches for automatic detection of sleep apnea/hypopnea based on human physiological signals: a review. Health Inf Sci Syst 2025; 13:7. [PMID: 39712669 PMCID: PMC11659556 DOI: 10.1007/s13755-024-00320-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/20/2024] [Indexed: 12/24/2024] Open
Abstract
Sleep apnea/hypopnea is a sleep disorder characterized by repeated pauses in breathing which could induce a series of health problems such as cardiovascular disease (CVD) and even sudden death. Polysomnography (PSG) is the most common way to diagnose sleep apnea/hypopnea. Considering that PSG data acquisition is complex and the diagnosis of sleep apnea/hypopnea requires manual scoring, it is very time-consuming and highly professional. With the development of wearable devices and AI techniques, more and more works have been focused on building machine and deep learning models that use single or multi-modal physiological signals to achieve automated detection of sleep apnea/hypopnea. This paper provides a comprehensive review of automatic sleep apnea/hypopnea detection methods based on AI-based techniques in recent years. We summarize the general process used by existing works with a flow chart, which mainly includes data acquisition, raw signal pre-processing, model construction, event classification, and evaluation, since few papers consider these. Additionally, the commonly used public database and pre-processing methods are also reviewed in this paper. After that, we separately summarize the existing methods related to different modal physiological signals including nasal airflow, pulse oxygen saturation (SpO2), electrocardiogram (ECG), electroencephalogram (EEG) and snoring sound. Furthermore, specific signal pre-processing methods based on the characteristics of different physiological signals are also covered. Finally, challenges need to be addressed, such as limited data availability, imbalanced data problem, multi-center study necessity etc., and future research directions related to AI are discussed.
Collapse
Affiliation(s)
- Dandan Peng
- The Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006 China
| | - Le Sun
- The Department of Jiangsu Collaborative Innovation Center of Atmospheric Environment and Equipment Technology (CICAEET), Nanjing University of Information Science and Technology, Nanjing, 210044 China
| | - Qian Zhou
- The School of Modern Posts, Nanjing University of Posts and Telecommunications, Nanjing, 210003 China
| | - Yanchun Zhang
- School of Computer Science, Zhejiang Normal University, Jinhua, 321000 China
- The Department of New Networks, Peng Cheng Laboratory, Shenzhen, 695571 China
| |
Collapse
|
4
|
Yang Y, Zhong Y, Chen L. EIciRNAs in focus: current understanding and future perspectives. RNA Biol 2025; 22:1-12. [PMID: 39711231 DOI: 10.1080/15476286.2024.2443876] [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] [Revised: 11/14/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024] Open
Abstract
Circular RNAs (circRNAs) are a unique class of covalently closed single-stranded RNA molecules that play diverse roles in normal physiology and pathology. Among the major types of circRNA, exon-intron circRNA (EIciRNA) distinguishes itself by its sequence composition and nuclear localization. Recent RNA-seq technologies and computational methods have facilitated the detection and characterization of EIciRNAs, with features like circRNA intron retention (CIR) and tissue-specificity being characterized. EIciRNAs have been identified to exert their functions via mechanisms such as regulating gene transcription, and the physiological relevance of EIciRNAs has been reported. Within this review, we present a summary of the current understanding of EIciRNAs, delving into their identification and molecular functions. Additionally, we emphasize factors regulating EIciRNA biogenesis and the physiological roles of EIciRNAs based on recent research. We also discuss the future challenges in EIciRNA exploration, underscoring the potential for novel functions and functional mechanisms of EIciRNAs for further investigation.
Collapse
Affiliation(s)
- Yan Yang
- Department of Cardiology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| | - Yinchun Zhong
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Liang Chen
- Department of Cardiology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
5
|
Meng Q, Tian L, Liu G, Zhang X. EEG-based cross-subject passive music pitch perception using deep learning models. Cogn Neurodyn 2025; 19:6. [PMID: 39758357 PMCID: PMC11699146 DOI: 10.1007/s11571-024-10196-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/22/2024] [Accepted: 11/06/2024] [Indexed: 01/07/2025] Open
Abstract
Pitch plays an essential role in music perception and forms the fundamental component of melodic interpretation. However, objectively detecting and decoding brain responses to musical pitch perception across subjects remains to be explored. In this study, we employed electroencephalography (EEG) as an objective measure to obtain the neural responses of musical pitch perception. The EEG signals from 34 subjects under hearing violin sounds at pitches G3 and B6 were collected with an efficient passive Go/No-Go paradigm. The lightweight modified EEGNet model was proposed for EEG-based pitch classification. Specifically, within-subject modeling with the modified EEGNet model was performed to construct individually optimized models. Subsequently, based on the within-subject model pool, a classifier ensemble (CE) method was adopted to construct the cross-subject model. Additionally, we analyzed the optimal time window of brain decoding for pitch perception in the EEG data and discussed the interpretability of these models. The experiment results show that the modified EEGNet model achieved an average classification accuracy of 77% for within-subject modeling, significantly outperforming other compared methods. Meanwhile, the proposed CE method achieved an average accuracy of 74% for cross-subject modeling, significantly exceeding the chance-level accuracy of 50%. Furthermore, we found that the optimal EEG data window for the pitch perception lies 0.4 to 0.9 s onset. These promising results demonstrate that the proposed methods can be effectively used in the objective assessment of pitch perception and have generalization ability in cross-subject modeling.
Collapse
Affiliation(s)
- Qiang Meng
- School of Integrated Circuits, Shandong University, 1500 Shunhua Road, Jinan, Shandong 250101 China
| | - Lan Tian
- School of Integrated Circuits, Shandong University, 1500 Shunhua Road, Jinan, Shandong 250101 China
| | - Guoyang Liu
- School of Integrated Circuits, Shandong University, 1500 Shunhua Road, Jinan, Shandong 250101 China
| | - Xue Zhang
- School of Integrated Circuits, Shandong University, 1500 Shunhua Road, Jinan, Shandong 250101 China
| |
Collapse
|
6
|
Wu HT, Liao CC, Peng CF, Lee TY, Liao PH. Exploring the application of machine learning to identify the correlations between phthalate esters and disease: enhancing nursing assessments. Health Inf Sci Syst 2025; 13:10. [PMID: 39736874 PMCID: PMC11683034 DOI: 10.1007/s13755-024-00324-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 12/10/2024] [Indexed: 01/01/2025] Open
Abstract
Background Health risks associated with phthalate esters depend on exposure level, individual sensitivities, and other contributing factors. Purpose This study employed artificial intelligence algorithms while applying data mining techniques to identify correlations between phthalate esters [di(2-ethylhexyl) phthalate, DEHP], lifestyle factors, and disease outcomes. Methods We conducted exploratory analysis using demographic and laboratory data collected from the Taiwan Biobank. The study developed a prediction model to examine the relationship between phthalate esters and the risk of developing certain diseases based on various artificial intelligence algorithms, including logistic regression, artificial neural networks, and Bayesian networks. Results The results indicate that phthalate esters exhibited a greater impact on bone and joint issues than heart problems. We observed that DEHP metabolites, such as mono(2-carboxymethylhexyl) phthalate, mono-n-butyl phthalate, and monoethylphthalate, leave higher residue in females than in males, with statistically significant differences. Monoethylphthalate levels were lower in individuals who exercised regularly than those who did not, indicating statistically significant differences. Conclusions This study's findings can serve as a valuable reference for clinical nursing assessments regarding diseases related to osteoporosis, arthritis, and musculoskeletal pain. Medical professionals can enhance care quality by considering factors beyond patients' essential physical assessment items.Trial Registration: This study was registered under NCT05892029 on May 5, 2023, retrospectively.
Collapse
Affiliation(s)
- Hao-Ting Wu
- Department of Nursing, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Chien-Chang Liao
- Department of Gastroenterologist, Tainan Municipal Hospital (Managed By Show Chwan Medical Care Corporation), Tainan, Taiwan
| | - Chiung-Fang Peng
- Department of Research, Taiwan Academy of Ecological Hazard & Health Management, Taipei, Taiwan
| | - Tso-Ying Lee
- Nursing Reserach center & School of Nursing, Taipei Medical University Hospital & Taipei Medical University, Taipei, Taiwan
| | - Pei-Hung Liao
- School of Nursing, National Taipei University of Nursing and Health Sciences, No. 365, Ming-Te Road, Peitou District, Taipei, 112 Taiwan
| |
Collapse
|
7
|
Wei S, Yang W, Wang E, Wang S, Li Y. A 3D decoupling Alzheimer's disease prediction network based on structural MRI. Health Inf Sci Syst 2025; 13:17. [PMID: 39846055 PMCID: PMC11748674 DOI: 10.1007/s13755-024-00333-3] [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: 02/27/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Purpose This paper aims to develop a three-dimensional (3D) Alzheimer's disease (AD) prediction method, thereby bettering current predictive methods, which struggle to fully harness the potential of structural magnetic resonance imaging (sMRI) data. Methods Traditional convolutional neural networks encounter pressing difficulties in accurately focusing on the AD lesion structure. To address this issue, a 3D decoupling, self-attention network for AD prediction is proposed. Firstly, a multi-scale decoupling block is designed to enhance the network's ability to extract fine-grained features by segregating convolutional channels. Subsequently, a self-attention block is constructed to extract and adaptively fuse features from three directions (sagittal, coronal and axial), so that more attention is geared towards brain lesion areas. Finally, a clustering loss function is introduced and combined with the cross-entropy loss to form a joint loss function for enhancing the network's ability to discriminate between different sample types. Results The accuracy of our model is 0.985 for the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset and 0.963 for the Australian Imaging, Biomarker & Lifestyle (AIBL) dataset, both of which are higher than the classification accuracy of similar tasks in this category. This demonstrates that our model can accurately distinguish between normal control (NC) and Alzheimer's Disease (AD), as well as between stable mild cognitive impairment (sMCI) and progressive mild cognitive impairment (pMCI). Conclusion The proposed AD prediction network exhibits competitive performance when compared with state-of-the-art methods. The proposed model successfully addresses the challenges of dealing with 3D sMRI image data and the limitations stemming from inadequate information in 2D sections, advancing the utility of predictive methods for AD diagnosis and treatment.
Collapse
Affiliation(s)
- Shicheng Wei
- School of Mathematics and Computing, University of Southern Queensland, 487-535 West Street, Toowoomba, QLD 4350 Australia
| | - Wencheng Yang
- School of Mathematics and Computing, University of Southern Queensland, 487-535 West Street, Toowoomba, QLD 4350 Australia
| | - Eugene Wang
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC Australia
| | - Song Wang
- Department of Engineering, La Trobe University, Bundoora, VIC 3086 Australia
| | - Yan Li
- School of Mathematics and Computing, University of Southern Queensland, 487-535 West Street, Toowoomba, QLD 4350 Australia
| |
Collapse
|
8
|
Ameen ZS, Mubarak AS, Hamad M, Hamoudi R, Jemimah S, Ozsahin DU, Hamad M. Incorporating time as a third dimension in transcriptomic analysis using machine learning and explainable AI. Comput Biol Chem 2025; 117:108432. [PMID: 40132403 DOI: 10.1016/j.compbiolchem.2025.108432] [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: 01/13/2025] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025]
Abstract
Transcriptomic data analysis entails the measurement of RNA transcript (gene expression products) abundance in a cell or a cell population at a single point in time. In other words, transcriptomics as it is currently practiced is two-dimensional (2DTA). Gene expression profiling by 2DTA has proven invaluable in furthering our understanding of numerous biological processes in health and disease. That said, shortcomings including technical variability, small sample size, differential rates of transcript decay, and the lack of linearity between transcript abundance and functionality or the formation of functional proteins limit the interpretive utility and generalizability of transcriptomic data. 2DTA utility may also be constrained by its reliance on RNA extracts obtained at a single time point. In other words, much like judging a movie by a single frame, 2DTA can only provide a snapshot of the transcriptome at time of RNA extraction. Whether this perceived "temporality" problem is real and whether it has any bearing on transcriptomic data interpretation have yet to be addressed. To investigate this problem, 25 publicly available datasets relating to MCF-7 cells, where RNA extracts obtained at 12- or 48-hours post-culture were subjected to transcriptomic analysis. The individual datasets were downloaded and compiled into two separate datasets (MCF-7 U12hr and MCF-7 U48hr). To comparatively analyze the two compiled datasets, three machine learning approaches (decision trees (DT), random forests (RF), and XGBoost (Extreme Gradient Boosting)) were used as classifiers to search for genes with distinct expression patterns between the two groups. Shapley additive explanation (SHAP), an explainable AI method, was used to assess the fundamental principles of the DT, RF, and XGBoost models. Coefficient of Determination (DC), Mean Absolute Error (MAE), and Mean Squared Error (MSE) were used to evaluate the models. The results show that the two datasets exhibited very significant gene expression patterns. The XGBoost model performed better than the DT or RF models with MSE, MAE, and DC values of 0.00028, 0.00028, and 0.95778 respectively. These observations suggest that time, as a third dimension, can impact transcriptomic data interpretation and that machine learning and explainable AI are useful tools in resolving the temporality problem in transcriptomics.
Collapse
Affiliation(s)
- Zubaida Said Ameen
- Operational Research Center in Healthcare, Near East University, Mersin 99138, Turkey
| | - Auwalu Saleh Mubarak
- Operational Research Center in Healthcare, Near East University, Mersin 99138, Turkey
| | - Mohamed Hamad
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, UAE; Research Institute of Medical and Health Sciences, University of Sharjah, UAE
| | - Rifat Hamoudi
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, UAE; BIMAI-Lab, Biomedically Informed Artificial Intelligence Laboratory, University of Sharjah, UAE; Division of Surgery and Interventional Science, University College London, London NW3 2QG, UK
| | - Sherlyn Jemimah
- Department of Biology, College of Science, American University of Sharjah, UAE
| | - Dilber Uzun Ozsahin
- Operational Research Center in Healthcare, Near East University, Mersin 99138, Turkey; Research Institute of Medical and Health Sciences, University of Sharjah, UAE; Department of Diagnostic Medical Imaging, College of Health Sciences, University of Sharjah, UAE.
| | - Mawieh Hamad
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, UAE; Research Institute of Medical and Health Sciences, University of Sharjah, UAE.
| |
Collapse
|
9
|
Ni J. Intelligent Recognition of Goji Berry Pests Using CNN With Multi-Graphic-Occlusion Data Augmentation and Multiple Attention Fusion Mechanisms. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2025; 118:e70060. [PMID: 40262026 DOI: 10.1002/arch.70060] [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: 02/24/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
Goji berry is an important economic crop, yet pest infestations pose a significant threat to its yield and quality. Traditional pest identification mainly relies on manual inspection by experts with specialized knowledge, which is subjective, time-consuming, and labor-intensive. To address these issues, this experiment proposes an improved convolutional neural network (CNN) for accurate identification of 17 types of goji berry pests. Firstly, the original data set is augmented using a multi-graph-occlusion data augmentation method. Subsequently, the augmented data set is imported into the improved CNN for training. Based on the original ResNet18 model, a new CNN, named GojiNet, is constructed by embedding multi-attention fusion modules at appropriate locations. Experimental results demonstrate that GojiNet achieves an average recognition accuracy of 95.35%, representing a 2.60% improvement over the ResNet18 network. Notably, compared to the original network, the training time of this model increases only slightly, while its size is reduced, and the recognition accuracy is enhanced. The experiment verifies the performance of the GojiNet model through a series of evaluation indicators. This study confirms the tremendous potential and application prospects of deep learning in pest identification, providing a referential solution for intelligent and precise pest identification.
Collapse
Affiliation(s)
- Jiangong Ni
- School of Electronics and Information Engineering, Hangzhou Dianzi University, Hangzhou, China
| |
Collapse
|
10
|
Zhao X, Zhang S, Zhang T, Cao Y, Liu J. A small-scale data driven and graph neural network based toxicity prediction method of compounds. Comput Biol Chem 2025; 117:108393. [PMID: 40048921 DOI: 10.1016/j.compbiolchem.2025.108393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 04/22/2025]
Abstract
Toxicity prediction is crucial in drug discovery, helping identify safe compounds and reduce development risks. However, the lack of known toxicity data for most compounds is a major challenge. Recently, data-driven models have gained attention as a more efficient alternative to traditional in vivo and in vitro experiments. In this paper, we propose a small-scale, data-driven toxicity prediction method based on Graph Neural Network (GNN). We introduce a joint learning strategy for multiple toxicity types and construct a graph-based model, JLGCN-MTT, to improve prediction accuracy. In addition, we integrate a transfer learning strategy that leverages data from multiple toxicity types, allowing the model to make reliable predictions even when data for a specific toxicity type is limited. We conducted experiments using data from 3566 compounds in the Tox21 dataset, which contains 12 types of toxicity-related bioactivity data. The experimental results show that JLGCN-MTT outperforms traditional machine learning methods and single-task GNN in all 12 toxicity prediction tasks, with AUC improving by over 10% in 11 tasks. For small-scale data with 50, 100, and 300 training samples, the AUC improved in all cases, with the highest improvement of 11% observed when the sample size was 50. These results demonstrate that the small-scale, data-driven toxicity prediction method we propose can achieve high prediction accuracy.
Collapse
Affiliation(s)
- Xin Zhao
- School of Electronic and Information Engineering, Tianjin University, 92 Weijin Road, Tianjin, 300072, Tianjin, China
| | - Shuyi Zhang
- School of Electronic and Information Engineering, Tianjin University, 92 Weijin Road, Tianjin, 300072, Tianjin, China
| | - Tao Zhang
- School of Electronic and Information Engineering, Tianjin University, 92 Weijin Road, Tianjin, 300072, Tianjin, China.
| | - Yahui Cao
- School of Electronic and Information Engineering, Tianjin University, 92 Weijin Road, Tianjin, 300072, Tianjin, China
| | - Jingjing Liu
- International Engineering Institute, Tianjin University, 92 Weijin Road, Tianjin, 300072, Tianjin, China
| |
Collapse
|
11
|
Xie J, Zhong S, Huang D, Shao W. PocketDTA: A pocket-based multimodal deep learning model for drug-target affinity prediction. Comput Biol Chem 2025; 117:108416. [PMID: 40073710 DOI: 10.1016/j.compbiolchem.2025.108416] [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: 11/05/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025]
Abstract
Drug-target affinity prediction is a fundamental task in the field of drug discovery. Extracting and integrating structural information from proteins effectively is crucial to enhance the accuracy and generalization of prediction, which remains a substantial challenge. This paper proposes a pocket-based multimodal deep learning model named PocketDTA for drug-target affinity prediction, based on the principle of "structure determines function". PocketDTA introduces the pocket graph structure that encodes protein residue features pretrained using a biological language model as nodes, while edges represent different protein sequences and spatial distances. This approach overcomes the limitations of lack of spatial information in traditional prediction models with only protein sequence input. Furthermore, PocketDTA employs relational graph convolutional networks at both atomic and residue levels to extract structural features from drugs and proteins. By integrating multimodal information through deep neural networks, PocketDTA combines sequence and structural data to improve affinity prediction accuracy. Experimental results demonstrate that PocketDTA outperforms state-of-the-art prediction models across multiple benchmark datasets by showing strong generalization under more realistic data splits and confirming the effectiveness of pocket-based methods for affinity prediction.
Collapse
Affiliation(s)
- Jiang Xie
- School of Computer Engineering and Science, Shanghai University, Shanghai, 200444, China
| | - Shengsheng Zhong
- School of Computer Engineering and Science, Shanghai University, Shanghai, 200444, China
| | - Dingkai Huang
- School of Computer Engineering and Science, Shanghai University, Shanghai, 200444, China
| | - Wei Shao
- Scientific Research Management Department, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
12
|
Lee S, Park JS, Hong JH, Woo H, Lee CH, Yoon JH, Lee KB, Chung S, Yoon DS, Lee JH. Artificial intelligence in bacterial diagnostics and antimicrobial susceptibility testing: Current advances and future prospects. Biosens Bioelectron 2025; 280:117399. [PMID: 40184880 DOI: 10.1016/j.bios.2025.117399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025]
Abstract
Recently, artificial intelligence (AI) has emerged as a transformative tool, enhancing the speed, accuracy, and scalability of bacterial diagnostics. This review explores the role of AI in revolutionizing bacterial detection and antimicrobial susceptibility testing (AST) by leveraging machine learning models, including Random Forest, Support Vector Machines (SVM), and deep learning architectures such as Convolutional Neural Networks (CNNs) and transformers. The integration of AI into these methods promises to address the current limitations of traditional techniques, offering a path toward more efficient, accessible, and reliable diagnostic solutions. In particular, AI-based approaches have demonstrated significant potential in resource-limited settings by enabling cost-effective and portable diagnostic solutions, reducing dependency on specialized infrastructure, and facilitating remote bacterial detection through smartphone-integrated platforms and telemedicine applications. This review highlights AI's transformative role in automating data analysis, minimizing human error, and delivering real-time diagnostic results, ultimately improving patient outcomes and optimizing healthcare efficiency. In addition, we not only examine the current advances in machine learning and deep learning but also review their applications in plate counting, mass spectrometry, morphology-based and motion-based microscopic detection, holographic microscopy, colorimetric and fluorescence detection, electrochemical sensors, Raman and Surface-Enhanced Raman Spectroscopy (SERS), and Atomic Force Microscopy (AFM) for bacterial diagnostics and AST. Finally, we discuss the future directions and potential advancements in AI-driven bacterial diagnostics.
Collapse
Affiliation(s)
- Seungmin Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; School of Biomedical Engineering, Korea University, 145 Anam-ro, Seongbuk, Seoul, 02841, Republic of Korea
| | - Jeong Soo Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; School of Mechanical Engineering, Korea University, 145 Anam-ro, Seoungbuk-gu, Seoul, 02841, Republic of Korea
| | - Ji Hye Hong
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; School of Biomedical Engineering, Korea University, 145 Anam-ro, Seongbuk, Seoul, 02841, Republic of Korea
| | - Hyowon Woo
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Chang-Hyun Lee
- Department of Electrical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon, Seoul, 01897, Republic of Korea
| | - Ju Hwan Yoon
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Department of Electrical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon, Seoul, 01897, Republic of Korea
| | - Ki-Baek Lee
- Department of Electrical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon, Seoul, 01897, Republic of Korea
| | - Seok Chung
- School of Mechanical Engineering, Korea University, 145 Anam-ro, Seoungbuk-gu, Seoul, 02841, Republic of Korea.
| | - Dae Sung Yoon
- School of Biomedical Engineering, Korea University, 145 Anam-ro, Seongbuk, Seoul, 02841, Republic of Korea; Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02841, Republic of Korea; Astrion Inc, Seoul, 02841, Republic of Korea.
| | - Jeong Hoon Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Department of Integrative Energy Engineering, College of Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| |
Collapse
|
13
|
Canavesi I, Viswakarma N, Epel B, McMillan A, Kotecha M. Accelerated EPR imaging using deep learning denoising. Magn Reson Med 2025; 94:436-446. [PMID: 40096518 DOI: 10.1002/mrm.30473] [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: 11/02/2024] [Revised: 01/15/2025] [Accepted: 02/05/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE Trityl OXO71-based pulse electron paramagnetic resonance imaging (EPRI) is an excellent technique to obtain partial pressure of oxygen (pO2) maps in tissues. In this study, we used deep learning techniques to denoise 3D EPR amplitude and pO2 maps. METHODS All experiments were performed using a 25 mT EPR imager, JIVA-25®. The MONAI implementation of four neural networks (autoencoder, Attention UNet, UNETR, and UNet) was tested, and the best model (UNet) was then enhanced with joint bilateral filters (JBF). The training dataset was comprised of 227 3D images (56 in vivo and 171 in vitro), 159 images for training, 45 for validation, and 23 for testing. UNet with 1, 2, and 3 JBF layers was tested to improve image SNR, focusing on multiscale structural similarity index measure and edge sensitivity preservation. The trained algorithm was tested using acquisitions with 15, 30, and 150 averages in vitro with a sealed deoxygenated OXO71 phantom and in vivo with fibrosarcoma tumors grown in a hind leg of C3H mice. RESULTS We demonstrate that UNet with 2 JBF layers (UNet+JBF2) provides the best outcome. We demonstrate that using the UNet+JBF2 model, the SNR of 15-shot amplitude maps provides higher SNR compared to 150-shot pre-filter maps, both in phantoms and in tumors, therefore, allowing 10-fold accelerated imaging. We demonstrate that the trained algorithm improves SNR in pO2 maps. CONCLUSIONS We demonstrate the application of deep learning techniques to EPRI denoising. Higher SNR will bring the EPRI technique one step closer to clinics.
Collapse
Affiliation(s)
- Irene Canavesi
- Oxygen Measurement Core, O2M Technologies, LLC, Chicago, Illinois, USA
| | - Navin Viswakarma
- Oxygen Measurement Core, O2M Technologies, LLC, Chicago, Illinois, USA
| | - Boris Epel
- Oxygen Measurement Core, O2M Technologies, LLC, Chicago, Illinois, USA
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois, USA
| | - Alan McMillan
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | |
Collapse
|
14
|
Kim J, Lee SJ, Jung D, Kim HY, Lee JI, Seo M, Kim S, Choi J, Yu WJ, Cho H. Development of a deep neural network model based on high throughput screening data for predicting synergistic estrogenic activity of binary mixtures for consumer products. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137650. [PMID: 40010213 DOI: 10.1016/j.jhazmat.2025.137650] [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: 10/28/2024] [Revised: 02/03/2025] [Accepted: 02/15/2025] [Indexed: 02/28/2025]
Abstract
A paradigm of chemical risk assessment is continuously extending from focusing on 'single substances' to more comprehensive approaches that examines the combined toxicity among different components in 'mixtures.' This change aims to account for the cocktail effect arising from the toxicological interactions in mixtures, which can lead to increased risks. More than 1000 potential endocrine-disrupting chemicals (EDCs) have been reported, and they can be included in different industrial and consumer chemical products and released to the environment as pollutants of emerging environmental concern. Although extensive studies involving both experiments and predictions have investigated individual EDCs, predictions of their synergistic effects are still relatively lacking, an area that requires further investigation. In this study, we extensively investigated substances in consumer products, mainly marketed in South Korea, that might exhibit estrogenic activity or reproductive toxicity. A high throughput screening (HTS) assay based on OECD Test Guideline 455 for hERαHeLa-9903 cells was constructed, and 435 substances were screened using the HTS. Thirty-five (potential) estrogenic agonists were selected, and their 1412 binary mixtures that could be prepared in four different ratios were systematically tested, considering the available effective concentrations of substances and the solubility of their resulting mixtures. The best empirical dose-response curves of 35 substances and 917 mixtures were derived in this study. Based on the HTS data, a deep neural network model was developed (area under the curve (AUC): 0.837-0.881) and compared with a random forest model (AUC: 0.656-0.829) to screen for the synergistic estrogenic activity of binary mixtures.
Collapse
Affiliation(s)
- Jongwoon Kim
- Digital Chemical Research Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea.
| | - Seung-Jin Lee
- Developmental and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Daeyoung Jung
- Center for Rare Disease Therapeutic Technology, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Hyun Young Kim
- Center for Rare Disease Therapeutic Technology, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Jung-In Lee
- Center for Rare Disease Therapeutic Technology, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Myungwon Seo
- Chemical Analysis Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Sunmi Kim
- Chemical Analysis Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Jiwon Choi
- Chemical Analysis Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Wook-Joon Yu
- Developmental and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea.
| | - Heeyeong Cho
- Center for Rare Disease Therapeutic Technology, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; Medicinal Chemistry and Pharmacology, University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
15
|
Luo X, Li B, Zhu R, Tai Y, Wang Z, He Q, Zhao Y, Bi X, Wu C. Development and validation of an interpretable machine learning model for predicting in-hospital mortality for ischemic stroke patients in ICU. Int J Med Inform 2025; 198:105874. [PMID: 40073651 DOI: 10.1016/j.ijmedinf.2025.105874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/12/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Timely and accurate outcome prediction is essential for clinical decision-making for ischemic stroke patients in the intensive care unit (ICU). However, the interpretation and translation of predictive models into clinical applications are equally crucial. This study aims to develop an interpretable machine learning (IML) model that effectively predicts in-hospital mortality for ischemic stroke patients. METHODS In this study, an IML model was developed and validated using multicenter cohorts of 3225 ischemic stroke patients admitted to the ICU. Nine machine learning (ML) models, including logistic regression (LR), K-nearest neighbors (KNN), naive Bayes (NB), decision tree (DT), support vector machine (SVM), random forest (RF), XGBoost, LightGBM, and artificial neural network (ANN), were developed to predict in-hospital mortality using data from the MIMIC-IV and externally validated in Shanghai Changhai Hospital. Feature selection was conducted using three algorithms. Model's performance was assessed using area under the receiver operating characteristic (AUROC), accuracy, sensitivity, specificity and F1 score. Calibration curve and Brier score were used to evaluate the degree of calibration of the model, and decision curve analysis were generated to assess the net clinical benefit. Additionally, the SHapley Additive exPlanations (SHAP) method was employed to evaluate the risk of in-hospital mortality among ischemic stroke patients admitted to the ICU. RESULTS Mechanical ventilation, age, statins, white blood cell, blood urea nitrogen, hematocrit, warfarin, bicarbonate and systolic blood pressure were selected as the nine most influential variables. The RF model demonstrated the most robust predictive performance, achieving AUROC values of 0.908 and 0.858 in the testing set and external validation set, respectively. Calibration curves also revealed a high consistency between observations and predictions. Decision curve analysis showed that the model had the greatest net benefit rate when the prediction probability threshold is 0.10 ∼ 0.80. SHAP was employed to interpret the RF model. In addition, we have developed an online prediction calculator for ischemic stroke patients. CONCLUSION This study develops a machine learning-based calculator to predict the probability of in-hospital mortality among patients with ischemic stroke in ICU. The calculator has the potential to guide clinical decision-making and improve the care of patients with ischemic stroke by identifying patients at a higher risk of in-hospital mortality.
Collapse
Affiliation(s)
- Xiao Luo
- Department of Military Health Statistics, Naval Medical University, Shanghai, China
| | - Binghan Li
- Department of Neurology, Changhai Hospital, the First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Ronghui Zhu
- Department of Military Health Statistics, Naval Medical University, Shanghai, China
| | - Yaoyong Tai
- Department of Military Health Statistics, Naval Medical University, Shanghai, China
| | - Zongyu Wang
- Department of Military Health Statistics, Naval Medical University, Shanghai, China; School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Qian He
- Department of Military Health Statistics, Naval Medical University, Shanghai, China
| | - Yanfang Zhao
- Department of Military Health Statistics, Naval Medical University, Shanghai, China
| | - Xiaoying Bi
- Department of Neurology, Changhai Hospital, the First Affiliated Hospital of Naval Medical University, Shanghai, China.
| | - Cheng Wu
- Department of Military Health Statistics, Naval Medical University, Shanghai, China.
| |
Collapse
|
16
|
Yadav MAP, Patil DS. "Optimizing sEMG Gesture Recognition with Stacked Autoencoder Neural Network for Bionic Hand". MethodsX 2025; 14:103207. [PMID: 40071216 PMCID: PMC11894319 DOI: 10.1016/j.mex.2025.103207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
This study presents a novel deep learning approach for surface electromyography (sEMG) gesture recognition using stacked autoencoder neural network (SAE)s. The method leverages hierarchical representation learning to extract meaningful features from raw sEMG signals, enhancing the precision and robustness of gesture classification.•Feature Extraction and Classification MODWT Decomposition: The sEMG signals were decomposed using the MODWT DECOMPOSITION(Maximal Overlap Discrete Wavelet Transform) to capture various frequency components.•Time Domain Parameters: A total of 28 features per subject were extracted from the time domain, including statistical and spectral features.•Classifier Evaluation: Initial evaluations involved Autoencoder and LDA (Linear Discriminant Analysis) classifiers, with Autoencoder achieving an average accuracy of 77.96 % ± 1.24, outperforming LDA's 65.36 % ± 1.09.Advanced Neural Network Approach: Stacked Autoencoder Neural Network: To address challenges in distinguishing similar gestures within grasp groups, a Stacked Autoencoder Neural Network was employed. This advanced neural network architecture improved classification accuracy to over 100 %, demonstrating its effectiveness in handling complex gesture recognition tasks. These findings emphasize the significant potential of deep learning models in enhancing prosthetic control and rehabilitation technologies. . To verify these findings, we developed a 3d hand module in ADAMS software that is simulated using Matlab-ADAMS cosimulation.
Collapse
Affiliation(s)
- Mr. Amol Pandurang Yadav
- All India Shri Shivaji Memorial Society's Institute Of Information Technology, India
- Bharati Vidyapeeth's College Of Engineering for Women, Pune, India
| | | |
Collapse
|
17
|
Gou M, Zhang H, Qian N, Zhang Y, Sun Z, Li G, Wang Z, Dai G. Deep learning radiomics analysis for prediction of survival in patients with unresectable gastric cancer receiving immunotherapy. Eur J Radiol Open 2025; 14:100626. [PMID: 39807092 PMCID: PMC11728962 DOI: 10.1016/j.ejro.2024.100626] [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: 10/28/2024] [Revised: 12/03/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
Objective Immunotherapy has become an option for the first-line therapy of advanced gastric cancer (GC), with improved survival. Our study aimed to investigate unresectable GC from an imaging perspective combined with clinicopathological variables to identify patients who were most likely to benefit from immunotherapy. Method Patients with unresectable GC who were consecutively treated with immunotherapy at two different medical centers of Chinese PLA General Hospital were included and divided into the training and validation cohorts, respectively. A deep learning neural network, using a multimodal ensemble approach based on CT imaging data before immunotherapy, was trained in the training cohort to predict survival, and an internal validation cohort was constructed to select the optimal ensemble model. Data from another cohort were used for external validation. The area under the receiver operating characteristic curve was analyzed to evaluate performance in predicting survival. Detailed clinicopathological data and peripheral blood prior to immunotherapy were collected for each patient. Univariate and multivariable logistic regression analysis of imaging models and clinicopathological variables was also applied to identify the independent predictors of survival. A nomogram based on multivariable logistic regression was constructed. Result A total of 79 GC patients in the training cohort and 97 patients in the external validation cohort were enrolled in this study. A multi-model ensemble approach was applied to train a model to predict the 1-year survival of GC patients. Compared to individual models, the ensemble model showed improvement in performance metrics in both the internal and external validation cohorts. There was a significant difference in overall survival (OS) among patients with different imaging models based on the optimum cutoff score of 0.5 (HR = 0.20, 95 % CI: 0.10-0.37, P < 0.001). Multivariate Cox regression analysis revealed that the imaging models, PD-L1 expression, and lung immune prognostic index were independent prognostic factors for OS. We combined these variables and built a nomogram. The calibration curves showed that the C-index of the nomogram was 0.85 and 0.78 in the training and validation cohorts. Conclusion The deep learning model in combination with several clinical factors showed predictive value for survival in patients with unresectable GC receiving immunotherapy.
Collapse
Affiliation(s)
- Miaomiao Gou
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Hongtao Zhang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Niansong Qian
- Department of Thoracic Oncology, The Eighth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Yong Zhang
- Department of Medical Oncology, The Second Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Zeyu Sun
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Guang Li
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Zhikuan Wang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Guanghai Dai
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| |
Collapse
|
18
|
Cao Y, Tian Y, Chen KE, Ma Y, Zhang Q, Liu W, Mu Z, Su S, Cao C. Specific glycomacropeptide detection via polyacrylamide gel electrophoresis with dual imaging and signal-fusion deep learning. Food Chem 2025; 476:143293. [PMID: 39986063 DOI: 10.1016/j.foodchem.2025.143293] [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: 05/27/2024] [Revised: 01/31/2025] [Accepted: 02/07/2025] [Indexed: 02/24/2025]
Abstract
Herein, we report a sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) method featuring dual imaging and signal-fusion deep learning for specific identification and analysis of glycomacropeptide (GMP) in milk sample. Conventional SDS-PAGE methods lack specificity because of the signle staining of protein bands, and the overlap between GMP and β-lactoglobulin (βLg). Our dual imaging method generated a pair of complementary detection signals by recruiting intrinsic fluorescence imaging (IFI) and silver staining. Comparing the IFI image with the staining image highlighted the presence of GMP and differentiated it from βLg. Additionally, we trained a signal-fusion deep learning model to improve the quantitative performance of our method. The model fused the features extracted from the paired detection signals (IFI and staining) and accurately classified them into different mixing ratios (proportion of GMP-containing whey in the sample), indicating the potential for quantitative analysis on the mixing ratios of GMP added into whey sample. The developed method has the merits of specificity, sensitivity and simplilcity, and has potential to analysis of protein/peptides with unique IFI properties in food safety, basic research and biopharming etc.
Collapse
Affiliation(s)
- Yiren Cao
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Youli Tian
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ke-Er Chen
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Yixin Ma
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qiang Zhang
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Weiwen Liu
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhishen Mu
- Inner Mongolia Enterprise Key Laboratory of Dairy Nutrition, Health & Safety, Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Huhhot 011500, China.
| | - Shengpeng Su
- Inner Mongolia Enterprise Key Laboratory of Dairy Nutrition, Health & Safety, Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Huhhot 011500, China.
| | - Chengxi Cao
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
19
|
Wang Q, Zhang S, Zeng D, Xie Z, Guo H, Zeng T, Fan FL. Don't fear peculiar activation functions: EUAF and beyond. Neural Netw 2025; 186:107258. [PMID: 39987712 DOI: 10.1016/j.neunet.2025.107258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/16/2024] [Accepted: 02/05/2025] [Indexed: 02/25/2025]
Abstract
In this paper, we propose a new super-expressive activation function called the Parametric Elementary Universal Activation Function (PEUAF). We demonstrate the effectiveness of PEUAF through systematic and comprehensive experiments on various industrial and image datasets, including CIFAR-10, Tiny-ImageNet, and ImageNet. The models utilizing PEUAF achieve the best performance across several baseline industrial datasets. Specifically, in image datasets, the models that incorporate mixed activation functions (with PEUAF) exhibit competitive test accuracy despite the low accuracy of models with only PEUAF. Moreover, we significantly generalize the family of super-expressive activation functions, whose existence has been demonstrated in several recent works by showing that any continuous function can be approximated to any desired accuracy by a fixed-size network with a specific super-expressive activation function. Specifically, our work addresses two major bottlenecks in impeding the development of super-expressive activation functions: the limited identification of super-expressive functions, which raises doubts about their broad applicability, and their often peculiar forms, which lead to skepticism regarding their scalability and practicality in real-world applications.
Collapse
Affiliation(s)
- Qianchao Wang
- Center of Mathematical Artificial Intelligence, Department of Mathematics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Shijun Zhang
- Department of Applied Mathematics, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region of China
| | - Dong Zeng
- Department of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Zhaoheng Xie
- Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing, China
| | - Hengtao Guo
- Independent Researcher, 708 6th Ave N, Seattle, WA 98109, United States of America
| | - Tieyong Zeng
- Center of Mathematical Artificial Intelligence, Department of Mathematics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Feng-Lei Fan
- Center of Mathematical Artificial Intelligence, Department of Mathematics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China.
| |
Collapse
|
20
|
Xu X, Wang C, Yi Q, Ye J, Kong X, Ashraf SQ, Dearn KD, Hajiyavand AM. MedBin: A lightweight End-to-End model-based method for medical waste management. WASTE MANAGEMENT (NEW YORK, N.Y.) 2025; 200:114742. [PMID: 40088805 DOI: 10.1016/j.wasman.2025.114742] [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: 11/05/2024] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
The surge in medical waste has highlighted the urgent need for cost-effective and advanced management solutions. In this paper, a novel medical waste management approach, "MedBin," is proposed for automated sorting, reusing, and recycling. A comprehensive medical waste dataset, "MedBin-Dataset" is established, comprising 2,119 original images spanning 36 categories, with samples captured in various backgrounds. The lightweight "MedBin-Net" model is introduced to enable detection and instance segmentation of medical waste, enhancing waste recognition capabilities. Experimental results demonstrate the effectiveness of the proposed approach, achieving an average precision of 0.91, recall of 0.97, and F1-score of 0.94 across all categories with just 2.51 M parameters (where M stands for million, i.e., 2.51 million parameters), 5.20G FLOPs (where G stands for billion, i.e., 5.20 billion floating-point operations per second), and 0.60 ms inference time. Additionally, the proposed method includes a World Health Organization (WHO) Guideline-Based Classifier that categorizes detected waste into 5 types, each with a corresponding disposal method, following WHO medical waste classification standards. The proposed method, along with the dedicated dataset, offers a promising solution that supports sustainable medical waste management and other related applications. To access the MedBin-Dataset samples, please visit https://universe.roboflow.com/uob-ylti8/medbin_dataset. The source code for MedBin-Net can be found at https://github.com/Wayne3918/MedbinNet.
Collapse
Affiliation(s)
- Xiazhen Xu
- Department of Mechanical Engineering, School of Engineering, University of Birmingham, Birmingham B15 2TT, UK
| | - Chenyang Wang
- Department of Mechanical Engineering, School of Engineering, University of Birmingham, Birmingham B15 2TT, UK
| | - Qiufeng Yi
- Department of Mechanical Engineering, School of Engineering, University of Birmingham, Birmingham B15 2TT, UK
| | - Jiaqi Ye
- Department of Mechanical Engineering, School of Engineering, University of Birmingham, Birmingham B15 2TT, UK
| | - Xiangfei Kong
- Department of Mechanical Engineering, School of Engineering, University of Birmingham, Birmingham B15 2TT, UK
| | - Shazad Q Ashraf
- Queen Elizabeth Hospital, Mindelsohn Way, Birmingham B15 2GW, UK
| | - Karl D Dearn
- Department of Mechanical Engineering, School of Engineering, University of Birmingham, Birmingham B15 2TT, UK
| | - Amir M Hajiyavand
- Department of Mechanical Engineering, School of Engineering, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
21
|
Serricchio L, Bocchi D, Chilin C, Marino R, Negri M, Cammarota C, Ricci-Tersenghi F. Daydreaming Hopfield Networks and their surprising effectiveness on correlated data. Neural Netw 2025; 186:107216. [PMID: 39985975 DOI: 10.1016/j.neunet.2025.107216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 02/24/2025]
Abstract
To improve the storage capacity of the Hopfield model, we develop a version of the dreaming algorithm that perpetually reinforces the patterns to be stored (as in the Hebb rule), and erases the spurious memories (as in dreaming algorithms). For this reason, we called it Daydreaming. Daydreaming is not destructive and it converges asymptotically to stationary retrieval maps. When trained on random uncorrelated examples, the model shows optimal performance in terms of the size of the basins of attraction of stored examples and the quality of reconstruction. We also train the Daydreaming algorithm on correlated data obtained via the random-features model and argue that it spontaneously exploits the correlations thus increasing even further the storage capacity and the size of the basins of attraction. Moreover, the Daydreaming algorithm is also able to stabilize the features hidden in the data. Finally, we test Daydreaming on the MNIST dataset and show that it still works surprisingly well, producing attractors that are close to unseen examples and class prototypes.
Collapse
Affiliation(s)
- Ludovica Serricchio
- Dipartimento di Fisica, Sapienza Università di Roma, Piazzale Aldo Moro 5, Rome, 00185, Italy; Center for Life Nano & Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy
| | - Dario Bocchi
- Dipartimento di Fisica, Sapienza Università di Roma, Piazzale Aldo Moro 5, Rome, 00185, Italy
| | - Claudio Chilin
- Departamento de Física Teórica, Universidad Complutense, Pl. de las Ciencias 1, Madrid, 28040, Spain
| | - Raffaele Marino
- Physics Department, Università degli Studi di Firenze, Via Sansone 1, Firenze, 50019, Italy
| | - Matteo Negri
- Dipartimento di Fisica, Sapienza Università di Roma, Piazzale Aldo Moro 5, Rome, 00185, Italy; CNR-Nanotec Rome unit, Piazzale Aldo Moro 5, Rome, 00185, Italy.
| | - Chiara Cammarota
- Dipartimento di Fisica, Sapienza Università di Roma, Piazzale Aldo Moro 5, Rome, 00185, Italy; INFN Sezione di Roma1, Piazzale Aldo Moro 5, Rome, 00185, Italy
| | - Federico Ricci-Tersenghi
- Dipartimento di Fisica, Sapienza Università di Roma, Piazzale Aldo Moro 5, Rome, 00185, Italy; CNR-Nanotec Rome unit, Piazzale Aldo Moro 5, Rome, 00185, Italy; INFN Sezione di Roma1, Piazzale Aldo Moro 5, Rome, 00185, Italy
| |
Collapse
|
22
|
Ning B, Zeng L, Fan K, Chen F. Low-speed impact localization of wind turbine blades with a single sensor utilizing multiscale feature fusion convolutional neural networks. ULTRASONICS 2025; 150:107598. [PMID: 39955861 DOI: 10.1016/j.ultras.2025.107598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/19/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Impact, which may occur during manufacturing, serving and maintaining, is a significant threat to in-service composite structures, e.g. wind turbine blades. It calls for developing a method for assessment and localization of impact. In this paper, a single-sensor impact localization method based on deep learning is proposed. Specifically, a multiscale feature fusion convolutional neural network is designed, which, in combination with a convolutional block attention module, adaptively extracts features from single-sensor signals to achieve accurate region-level source localization. Complete ensemble empirical mode decomposition with adaptive noise is employed to reduce noise and extract intrinsic mode functions from acoustic emission signals, enabling more effective feature extraction. The decomposed signals are then converted into grayscale images, forming a dataset for the deep learning model. This approach allows for the extraction of rich feature information. A steel ball drop experiment is conducted to simulate the low-speed impact response of the wind turbine blade spar. The experimental results show significant advantages in localization accuracy. This study offers a promising solution for acoustic emission source region localization in complex composite structures.
Collapse
Affiliation(s)
- Botao Ning
- School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi Province 710049, China.
| | - Liang Zeng
- School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi Province 710049, China.
| | - Kaidi Fan
- School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi Province 710049, China.
| | - Feiyu Chen
- 713th Research Institute of China Shipbuilding Industry Corporation, Zhengzhou, Henan Province 450015, China.
| |
Collapse
|
23
|
Agboka KM, Abdel-Rahman EM, Salifu D, Kanji B, Ndjomatchoua FT, Guimapi RA, Ekesi S, Tobias L. Towards combining self-organizing maps (SOM) and convolutional neural network (CNN) for improving model accuracy: Application to malaria vectors phenotypic resistance. MethodsX 2025; 14:103198. [PMID: 39991436 PMCID: PMC11847465 DOI: 10.1016/j.mex.2025.103198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/29/2025] [Indexed: 02/25/2025] Open
Abstract
This study introduces a hybrid approach that combines unsupervised self-organizing maps (SOM) with a supervised convolutional neural network (CNN) to enhance model accuracy in vector-borne disease modeling. We applied this method to predict insecticide resistance (IR) status in key malaria vectors across Africa. Our results show that the combined SOM/CNN approach is more robust than a standalone CNN model, achieving higher overall accuracy and Kappa scores among others. This confirms the potential of the SOM/CNN hybrid as an effective and reliable tool for improving model accuracy in public health applications.•The hybrid model, combining SOM and CNN, was implemented to predict IR status in malaria vectors, providing enhanced accuracy across various validation metrics.•Results indicate a notable improvement in robustness and predictive accuracy over traditional CNN models.•The combined SOM/CNN approach demonstrated higher Kappa scores and overall model accuracy.
Collapse
Affiliation(s)
- Komi Mensah Agboka
- International Centre of Insect Physiology and Ecology (ICIPE), P.O. Box 30772 00100, Kenya
| | - Elfatih M. Abdel-Rahman
- International Centre of Insect Physiology and Ecology (ICIPE), P.O. Box 30772 00100, Kenya
- School of Agricultural, Earth, and Environmental Sciences, University of KwaZulu-Natal, Pietermaritzburg 3209, South Africa
| | - Daisy Salifu
- International Centre of Insect Physiology and Ecology (ICIPE), P.O. Box 30772 00100, Kenya
| | - Brian Kanji
- International Centre of Insect Physiology and Ecology (ICIPE), P.O. Box 30772 00100, Kenya
| | - Frank T. Ndjomatchoua
- Department of Plant Sciences, School of the Biological Sciences, University of Cambridge, Cambridge CB2 3EA, United Kingdom
| | - Ritter A.Y. Guimapi
- Biotechnology and Plant Health Division, Norwegian Institute of Bioeconomy Research (NIBIO), P.O. Box 115, Ås NO-1431, Norway
| | - Sunday Ekesi
- International Centre of Insect Physiology and Ecology (ICIPE), P.O. Box 30772 00100, Kenya
| | - Landmann Tobias
- International Centre of Insect Physiology and Ecology (ICIPE), P.O. Box 30772 00100, Kenya
| |
Collapse
|
24
|
Zhang A, Langenkamp M, Kleiman-Weiner M, Oikarinen T, Cushman F. Similar failures of consideration arise in human and machine planning. Cognition 2025; 259:106108. [PMID: 40086083 DOI: 10.1016/j.cognition.2025.106108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 02/25/2025] [Accepted: 03/01/2025] [Indexed: 03/16/2025]
Abstract
Humans are remarkably efficient at decision making, even in "open-ended" problems where the set of possible actions is too large for exhaustive evaluation. Our success relies, in part, on processes for calling to mind the right candidate actions. When these processes fail, the result is a kind of puzzle in which the value of a solution would be obvious once it is considered, but never gets considered in the first place. Recently, machine learning (ML) architectures have attained or even exceeded human performance on open-ended decision making tasks such as playing chess and Go. We ask whether the broad architectural principles that underlie ML success in these domains generate similar consideration failures to those observed in humans. We demonstrate a case in which they do, illuminating how humans make open-ended decisions, how this relates to ML approaches to similar problems, and how both architectures lead to characteristic patterns of success and failure.
Collapse
Affiliation(s)
- Alice Zhang
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, United States of America; Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, United States of America.
| | - Max Langenkamp
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, United States of America.
| | - Max Kleiman-Weiner
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, United States of America.
| | - Tuomas Oikarinen
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, United States of America.
| | - Fiery Cushman
- Department of Psychology, Harvard University, Cambridge, MA 02139, United States of America.
| |
Collapse
|
25
|
Galán J, González M, Moral P, García-Martín Á, Martínez JM. Transforming urban waste collection inventory: AI-Based container classification and Re-Identification. WASTE MANAGEMENT (NEW YORK, N.Y.) 2025; 199:25-35. [PMID: 40081303 DOI: 10.1016/j.wasman.2025.02.051] [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: 05/31/2024] [Revised: 02/05/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025]
Abstract
This work lays the groundwork for creating an automated system for the inventory of urban waste elements. Our primary contribution is the development of, to the best of our knowledge, the first re-identification system for urban waste elements that uses Artificial Intelligence and Computer Vision, incorporating information from a classification module and geolocation context to enhance post-processing performance. This re-identification system helps to create and update inventories by determining if a new image matches an existing element in the inventory based on visual similarity or, if not, by adding it as a new identity (new class or new identity of an existing class). Such a system could be highly valuable to local authorities and waste management companies, offering improved facility maintenance, geolocation, and additional applications. This work also addresses the dynamic nature of urban environments and waste management elements by exploring Continual Learning strategies to adapt pretrained systems to new settings with different urban elements. Experimental results show that the proposed system operates effectively across various container types and city layouts. These findings were validated through testing in two different Spanish locations, a "City" and a "Campus", differing in size, illumination conditions, seasons, urban design and container appearance. For the final re-identification system, the baseline system achieves 53.18 mAP (mean Average Precision) in the simple scenario, compared to 21.54 mAP in the complex scenario, with additional challenging unseen variability. Incorporating the proposed post-processing techniques significantly improved results, reaching 74.14 mAP and 71.75 mAP in the simple and complex scenario respectively.
Collapse
Affiliation(s)
- Javier Galán
- Video Processing and Understanding Lab, Universidad Aut́onoma de Madrid 28049 Madrid, Spain
| | - Miguel González
- Video Processing and Understanding Lab, Universidad Aut́onoma de Madrid 28049 Madrid, Spain
| | - Paula Moral
- Video Processing and Understanding Lab, Universidad Aut́onoma de Madrid 28049 Madrid, Spain
| | - Álvaro García-Martín
- Video Processing and Understanding Lab, Universidad Aut́onoma de Madrid 28049 Madrid, Spain.
| | - José M Martínez
- Video Processing and Understanding Lab, Universidad Aut́onoma de Madrid 28049 Madrid, Spain
| |
Collapse
|
26
|
Ljubimov VA, Sun T, Wang J, Li L, Wang PZ, Ljubimov AV, Holler E, Black KL, Kopeček J, Ljubimova JY, Yang J. Blood-brain barrier crossing biopolymer targeting c-Myc and anti-PD-1 activate primary brain lymphoma immunity: Artificial intelligence analysis. J Control Release 2025; 381:113611. [PMID: 40088978 DOI: 10.1016/j.jconrel.2025.113611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
Primary Central Nervous System Lymphoma is an aggressive central nervous system neoplasm with poor response to pharmacological treatment, partially due to insufficient drug delivery across blood-brain barrier. In this study, we developed a novel therapy for this lymphoma by combining a targeted nanopolymer treatment with an immune checkpoint inhibitor antibody (anti-PD-1). A N-(2-hydroxypropyl)methacrylamide copolymer-based nanoconjugate was designed to block tumor cell c-Myc oncogene expression by antisense oligonucleotide. Angiopep-2 peptide was conjugated to the copolymer to facilitate nanodrug crossing of the blood-brain barrier. Systemically administered polymeric nanodrug, alone or in combination with immune checkpoint inhibitor antibody anti-PD-1, was tested in syngeneic mouse model of A20 intracranial brain lymphoma. There was no significant survival difference between saline- and free anti-PD-1-treated groups. However, significant survival advantage vs. saline was observed upon treatment with nanodrug bearing Angiopep-2, H6 (6 histidines for endosome escape), and c-Myc antisense alone and especially when it was combined with anti-PD-1 antibody. Animal survival after combined treatment was also significantly increased vs. free anti-PD-1. Artificial Intelligence-assisted analysis of gene expression database after RNA-seq of tumors was used to find novel immune pathways, molecular targets and the most effective multifunctional drugs together with future drug prediction for brain lymphoma in vivo model. Spectral flow cytometry and RNA-seq analysis revealed a robust activation of tumor infiltrating T lymphocytes with enhanced interferon γ signaling and polarization to M1-type macrophages in treated tumors, which was confirmed by immunofluorescence staining. In summary, a new effective blood-brain barrier crossing nano immuno therapeutic system was developed that effectively blocked tumor c-Myc acting in combination with immune checkpoint inhibitor anti-PD-1 to treat primary brain lymphoma. The treatment improved survival of tumor-bearing animals through activation of both the adaptive and innate immune responses.
Collapse
Affiliation(s)
- Vladimir A Ljubimov
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, United States
| | - Tao Sun
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, United States
| | - Jiawei Wang
- Department of Molecular Pharmaceutics/CCCD, University of Utah, 20 S 2030 E, Salt Lake City, UT 84112, United States
| | - Lian Li
- Department of Molecular Pharmaceutics/CCCD, University of Utah, 20 S 2030 E, Salt Lake City, UT 84112, United States
| | - Paul Z Wang
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Alexander V Ljubimov
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, United States; Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States; Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Eggehard Holler
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, United States; Institut für Biophysik und Physikalische Biochemie Universität Regensburg, D-93040 Regensburg, Germany
| | - Keith L Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, United States; Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Jindřich Kopeček
- Department of Molecular Pharmaceutics/CCCD, University of Utah, 20 S 2030 E, Salt Lake City, UT 84112, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, United States
| | - Julia Y Ljubimova
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, United States.
| | - Jiyuan Yang
- Department of Molecular Pharmaceutics/CCCD, University of Utah, 20 S 2030 E, Salt Lake City, UT 84112, United States.
| |
Collapse
|
27
|
Lv W, Jia X, Tang B, Ma C, Fang X, Jin X, Niu Z, Han X. In silico modeling of targeted protein degradation. Eur J Med Chem 2025; 289:117432. [PMID: 40015161 DOI: 10.1016/j.ejmech.2025.117432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
Targeted protein degradation (TPD) techniques, particularly proteolysis-targeting chimeras (PROTAC) and molecular glue degraders (MGD), have offered novel strategies in drug discovery. With rapid advancement of computer-aided drug design (CADD) and artificial intelligence-driven drug discovery (AIDD) in the biomedical field, a major focus has become how to effectively integrate these technologies into the TPD drug discovery pipeline to accelerate development, shorten timelines, and reduce costs. Currently, the main research directions for applying CADD and AIDD in TPD include: 1) ternary complex modeling; 2) linker generation; 3) strategies to predict degrader targets, activities and ADME/T properties; 4) In silico degrader design and discovery. Models developed in these areas play a crucial role in target identification, drug design, and optimization at various stages of the discovery process. However, the limited size and quality of datasets related to TPD present challenges, leaving room for further improvement in these models. TPD involves the complex ubiquitin-proteasome system, with numerous factors influencing outcomes. Most current models adopt a static perspective to interpret and predict relevant tasks. In the future, it may be necessary to shift toward dynamic approaches that better capture the intricate relationships among these components. Furthermore, incorporating new and diverse chemical spaces will enhance the precision design and application of TPD agents.
Collapse
Affiliation(s)
- Wenxing Lv
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China; Hangzhou Institute of Advanced Technology, Hangzhou, 310000, China.
| | - Xiaojuan Jia
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China.
| | - Bowen Tang
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, China; Guangzhou New Block Technology Co., Ltd., Guangzhou, 510000, China.
| | - Chao Ma
- Guangzhou New Block Technology Co., Ltd., Guangzhou, 510000, China.
| | - Xiaopeng Fang
- Hangzhou Institute of Advanced Technology, Hangzhou, 310000, China.
| | - Xurui Jin
- MindRank AI, Hangzhou, 310000, China.
| | - Zhangming Niu
- MindRank AI, Hangzhou, 310000, China; National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK.
| | - Xin Han
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Guangxi Normal University), Guilin, 541004, China.
| |
Collapse
|
28
|
Shi MH, Zhang SW, Zhang QQ, Han Y, Zhang S. PLAGCA: Predicting protein-ligand binding affinity with the graph cross-attention mechanism. J Biomed Inform 2025; 165:104816. [PMID: 40139623 DOI: 10.1016/j.jbi.2025.104816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
Accurate prediction of protein-ligand binding affinity plays a crucial role in drug discovery. However, determining the binding affinity of protein-ligands through biological experimental approaches is both time-consuming and expensive. Although some computational methods have been developed to predict protein-ligands binding affinity, most existing methods extract the global features of proteins and ligands through separate encoders, without considering to extract the local pocket interaction features of protein-ligand complexes, resulting in the limited prediction accuracy. In this work, we proposed a novel Protein-Ligand binding Affinity prediction method (named PLAGCA) by introducing Graph Cross-Attention mechanism to learn the local three-dimensional (3D) features of protein-ligand pockets, and integrating the global sequence/string features and local graph interaction features of protein-ligand complexes. PLAGCA uses sequence encoding and self-attention to extract the protein/ligand global features from protein FASTA sequences/ligand SMILES strings, adopts graph neural network and cross-attention to extract the protein-ligand local interaction features from the molecular structures of protein binding pockets and ligands. All these features are concatenated and input into a multi-layer perceptron (MLP) for predicting the protein-ligand binding affinity. The experimental results show that our PLAGCA outperforms other state-of-the-art computational methods, and it can effectively predict protein-ligand binding affinity with superior generalization capability. PLAGCA can capture the critical functional residues that are important contribution to the protein-ligand binding.
Collapse
Affiliation(s)
- Ming-Hui Shi
- MOE Key Laboratory of Information Fusion Technology, School of Automation, Northwestern Polytechnical University, Xian 710072, China.
| | - Shao-Wu Zhang
- MOE Key Laboratory of Information Fusion Technology, School of Automation, Northwestern Polytechnical University, Xian 710072, China.
| | - Qing-Qing Zhang
- MOE Key Laboratory of Information Fusion Technology, School of Automation, Northwestern Polytechnical University, Xian 710072, China
| | - Yong Han
- MOE Key Laboratory of Information Fusion Technology, School of Automation, Northwestern Polytechnical University, Xian 710072, China
| | - Shanwen Zhang
- School of Computing, Xijing University, Xi'an, 710123, China
| |
Collapse
|
29
|
Teschendorff AE, Horvath S. Epigenetic ageing clocks: statistical methods and emerging computational challenges. Nat Rev Genet 2025; 26:350-368. [PMID: 39806006 DOI: 10.1038/s41576-024-00807-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2024] [Indexed: 01/16/2025]
Abstract
Over the past decade, epigenetic clocks have emerged as powerful machine learning tools, not only to estimate chronological and biological age but also to assess the efficacy of anti-ageing, cellular rejuvenation and disease-preventive interventions. However, many computational and statistical challenges remain that limit our understanding, interpretation and application of epigenetic clocks. Here, we review these computational challenges, focusing on interpretation, cell-type heterogeneity and emerging single-cell methods, aiming to provide guidelines for the rigorous construction of interpretable epigenetic clocks at cell-type and single-cell resolution.
Collapse
Affiliation(s)
- Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | | |
Collapse
|
30
|
Taori S, Lim S. Classification algorithms trained on simple (symmetric) lifting data perform poorly in predicting hand loads during complex (free-dynamic) lifting tasks. APPLIED ERGONOMICS 2025; 125:104427. [PMID: 39662372 DOI: 10.1016/j.apergo.2024.104427] [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: 05/24/2024] [Revised: 09/25/2024] [Accepted: 11/27/2024] [Indexed: 12/13/2024]
Abstract
The performance of machine learning (ML) algorithms is dependent on which dataset it has been trained on. While ML algorithms are increasingly used for lift risk assessment, many algorithms are often trained and tested on controlled simulation datasets, lacking the diversity of the lifting conditions. Consequently, concerns arise regarding their applicability in real-world scenarios characterized by substantial variations in lifting scenarios and postures. Our study investigates the impact of different lifting scenarios on the performance of ML algorithms trained on surface electromyography (sEMG) armband sensor data to classify hand-load levels (2.3 and 6.8 kg). Twelve healthy participants (6 male and 6 female) performed repetitive lifting tasks employing various lifting scenarios, including symmetric (S), asymmetric (A), and free-dynamic (F) techniques. Separate algorithms were developed using diverse training datasets (S, A, S+A, and F), ML classifiers, and sEMG features, and tested using the F dataset, representing unconstrained and naturalistic lifts. The mean accuracy and sensitivity were significantly lower in models trained on constrained (S) datasets compared to those trained on naturalistic lifts (F). The accuracy, precision, and sensitivity of models trained with frequency-domain sEMG features were greater than those trained with the time-domain features. In conclusion, ML algorithms trained on controlled symmetric lifts showed poor performance in predicting loads for dynamic, unconstrained lifts; thus, particular attention is needed when using such algorithms in real-world scenarios.
Collapse
Affiliation(s)
- Sakshi Taori
- Department of Industrial and Systems Engineering, Virginia Polytechnic Institute and State University, 1145 Perry Street, Blacksburg, VA, USA.
| | - Sol Lim
- Department of Industrial and Systems Engineering, Virginia Polytechnic Institute and State University, 1145 Perry Street, Blacksburg, VA, USA.
| |
Collapse
|
31
|
Mohan J, Sivasubramanian A, V S, Ravi V. Enhancing skin disease classification leveraging transformer-based deep learning architectures and explainable AI. Comput Biol Med 2025; 190:110007. [PMID: 40117795 DOI: 10.1016/j.compbiomed.2025.110007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/27/2025] [Accepted: 03/05/2025] [Indexed: 03/23/2025]
Abstract
Skin diseases affect over a third of the global population, yet their impact is often underestimated. Automating the classification of these diseases is essential for supporting timely and accurate diagnoses. This study leverages Vision Transformers, Swin Transformers, and DinoV2, introducing DinoV2 for the first time in dermatology tasks. On a 31-class skin disease dataset, DinoV2 achieves state-of-the-art results with a test accuracy of 96.48 ± 0.0138% and an F1-Score of 97.27%, marking a nearly 10% improvement over existing benchmarks. The robustness of DinoV2 is further validated on the HAM10000 and Dermnet datasets, where it consistently surpasses prior models. Comparative analysis also includes ConvNeXt and other CNN architectures, underscoring the benefits of transformer models. Additionally, explainable AI techniques like GradCAM and SHAP provide global heatmaps and pixel-level correlation plots, offering detailed insights into disease localization. These complementary approaches enhance model transparency and support clinical correlations, assisting dermatologists in accurate diagnosis and treatment planning. This combination of high performance and clinical relevance highlights the potential of transformers, particularly DinoV2, in dermatological applications.
Collapse
Affiliation(s)
- Jayanth Mohan
- Amrita School of Artificial Intelligence, Coimbatore, Amrita Vishwa Vidyapeetham, India.
| | - Arrun Sivasubramanian
- Amrita School of Artificial Intelligence, Coimbatore, Amrita Vishwa Vidyapeetham, India.
| | - Sowmya V
- Amrita School of Artificial Intelligence, Coimbatore, Amrita Vishwa Vidyapeetham, India.
| | - Vinayakumar Ravi
- Center for Artificial Intelligence, Prince Mohammad Bin Fahd University, Khobar, Saudi Arabia.
| |
Collapse
|
32
|
Zhou ZF, Huang D, Wang CD. Pyramid contrastive learning for clustering. Neural Netw 2025; 185:107217. [PMID: 39919524 DOI: 10.1016/j.neunet.2025.107217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/01/2025] [Accepted: 01/26/2025] [Indexed: 02/09/2025]
Abstract
With its ability of joint representation learning and clustering via deep neural networks, the deep clustering have gained significant attention in recent years. Despite the considerable progress, most of the previous deep clustering methods still suffer from three critical limitations. First, they tend to associate some distribution-based clustering loss to the neural network, which often overlook the sample-wise contrastiveness for discriminative representation learning. Second, they generally utilize the features learned at a single layer for the clustering process, which, surprisingly, cannot go beyond a single layer to explore multiple layers for joint multi-layer (multi-stage) learning. Third, they typically use the convolutional neural network (CNN) for clustering images, which focus on local information yet cannot well capture the global dependencies. To tackle these issues, this paper presents a new deep clustering method called pyramid contrastive learning for clustering (PCLC), which is able to incorporate a pyramidal contrastive architecture to jointly enforce contrastive learning and clustering at multiple network layers (or stages). Particularly, for an input image, two types of augmentations are first performed to generate two paralleled augmented views. To bridge the gap between the CNN (for capturing local information) and the Transformer (for reflecting global dependencies), a mixed CNN-Transformer based encoder is utilized as the backbone, whose CNN-Transformer blocks are further divided into four stages, thus giving rise to a pyramid of multi-stage feature representations. Thereafter, multiple stages of twin contrastive learning are simultaneously conducted at both the instance-level and the cluster-level, through the optimization of which the final clustering can be achieved. Extensive experiments on multiple challenging image datasets demonstrate the superior clustering performance of PCLC over the state-of-the-art. The source code is available at https://github.com/Zachary-Chow/PCLC.
Collapse
Affiliation(s)
- Zi-Feng Zhou
- College of Mathematics and Informatics, South China Agricultural University, Guangzhou, China.
| | - Dong Huang
- College of Mathematics and Informatics, South China Agricultural University, Guangzhou, China; Key Laboratory of Smart Agricultural Technology in Tropical South China, Ministry of Agriculture and Rural Affairs, China.
| | - Chang-Dong Wang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, China; Guangdong Key Laboratory of Big Data Analysis and Processing, Guangzhou, China.
| |
Collapse
|
33
|
Khaffafi B, Khoshakhalgh H, Keyhanazar M, Mostafapour E. Automatic cerebral microbleeds detection from MR images via multi-channel and multi-scale CNNs. Comput Biol Med 2025; 189:109938. [PMID: 40056835 DOI: 10.1016/j.compbiomed.2025.109938] [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: 11/06/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Computer-aided detection (CAD) systems have been widely used to assist medical professionals in interpreting medical images, aiding in the detection of potential diseases. Despite their usefulness, CAD systems cannot yet fully replace doctors in diagnosing many conditions due to limitations in current algorithms. Cerebral microbleeds (CMBs) are a critical area of concern for neurological health, and accurate detection of CMBs is essential for understanding their impact on brain function. This study aims to improve CMB detection by enhancing existing machine learning algorithms. METHODS This paper presents four CNN-based algorithms designed to enhance CMB detection. The detection methods are categorized into traditional machine learning approaches and deep learning-based methods. The traditional methods, while computationally efficient, offer lower sensitivity, while CNN-based approaches promise greater accuracy. The algorithms proposed in this study include a multi-channel CNN with optimized architecture and a multiscale CNN structure, both of which were designed to reduce false positives and improve overall performance. RESULTS The first CNN algorithm, with an optimized multi-channel architecture, demonstrated a sensitivity of 99.6 %, specificity of 99.3 %, and accuracy of 99.5 %. The fourth algorithm, based on a stable multiscale CNN structure, achieved sensitivity of 98.2 %, specificity of 97.4 %, and accuracy of 97.8 %. Both algorithms exhibited a significant reduction in false positives compared to traditional methods. The experiments conducted confirm the effectiveness of these algorithms in improving the precision and reliability of CMB detection. CONCLUSION The proposed CNN-based algorithms demonstrate a significant advancement in the automated detection of CMBs, with notable improvements in sensitivity, specificity, and accuracy. These results underscore the potential of deep learning models, particularly CNNs, in enhancing CAD systems for neurological disease detection and reducing diagnostic errors. Further research and optimization may allow these algorithms to be integrated into clinical practices, providing more reliable support for healthcare professionals.
Collapse
Affiliation(s)
- Behrang Khaffafi
- Department of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hadi Khoshakhalgh
- Department of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Mohammad Keyhanazar
- Department of Electrical and Computer Engineering, Urmia Branch, Islamic Azad University, Urmia, Iran.
| | - Ehsan Mostafapour
- Department of Electrical and Computer Engineering, Urmia University, Urmia, Iran.
| |
Collapse
|
34
|
Carboni L, Nwaigwe D, Mainsant M, Bayle R, Reyboz M, Mermillod M, Dojat M, Achard S. Exploring continual learning strategies in artificial neural networks through graph-based analysis of connectivity: Insights from a brain-inspired perspective. Neural Netw 2025; 185:107125. [PMID: 39847940 DOI: 10.1016/j.neunet.2025.107125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/24/2024] [Accepted: 01/02/2025] [Indexed: 01/25/2025]
Abstract
Artificial Neural Networks (ANNs) aim at mimicking information processing in biological networks. In cognitive neuroscience, graph modeling is a powerful framework widely used to study brain structural and functional connectivity. Yet, the extension of graph modeling to ANNs has been poorly explored especially in terms of functional connectivity (i.e. the contextual change of the activity's units in networks). In the perspective of designing more robust and interpretable ANNs, we study how a brain-inspired graph-based approach can be extended and used to investigate ANN properties and behaviors. We focus our study on different continual learning strategies inspired by the biological mechanisms and modeled with ANNs. We show that graph modeling offers a simple and elegant framework to deeply investigate ANNs, compare their performances, and explore deleterious behaviors such as catastrophic forgetting.
Collapse
Affiliation(s)
- Lucrezia Carboni
- Univ. Grenoble Alpes, CNRS, Inria, Grenoble INP, LJK, 38000 Grenoble, France; Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, GIN, 38000 Grenoble, France
| | - Dwight Nwaigwe
- Univ. Grenoble Alpes, CNRS, Inria, Grenoble INP, LJK, 38000 Grenoble, France; Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, GIN, 38000 Grenoble, France
| | - Marion Mainsant
- Univ. Grenoble Alpes, Univ. Savoie Mont Blanc, CNRS, LPNC, 38000 Grenoble, France; Univ. Grenoble Alpes, CEA, LIST, 38000 Grenoble, France
| | - Raphael Bayle
- Univ. Grenoble Alpes, CEA, LIST, 38000 Grenoble, France
| | - Marina Reyboz
- Univ. Grenoble Alpes, CEA, LIST, 38000 Grenoble, France
| | - Martial Mermillod
- Univ. Grenoble Alpes, Univ. Savoie Mont Blanc, CNRS, LPNC, 38000 Grenoble, France
| | - Michel Dojat
- Univ. Grenoble Alpes, CNRS, Inria, Grenoble INP, LJK, 38000 Grenoble, France; Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, GIN, 38000 Grenoble, France.
| | - Sophie Achard
- Univ. Grenoble Alpes, CNRS, Inria, Grenoble INP, LJK, 38000 Grenoble, France
| |
Collapse
|
35
|
Debsarkar SS, Aronow B, Prasath VBS. Advancements in automated nuclei segmentation for histopathology using you only look once-driven approaches: A systematic review. Comput Biol Med 2025; 190:110072. [PMID: 40138968 DOI: 10.1016/j.compbiomed.2025.110072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/05/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Histopathology image analysis plays a pivotal role in disease diagnosis and treatment planning, relying heavily on accurate nuclei segmentation for extracting vital cellular information. In recent years, artificial intelligence (AI) and in particular deep learning models have been applied successfully in solving computational pathology image analysis tasks. The You Only Look Once (YOLO) object detection framework, which is based on a convolutional neural network (CNN) architecture has gained traction across various domains for its real-time processing capabilities. This systematic review aims to comprehensively explore and evaluate the advancements, challenges, and applications of YOLO-based methodologies in nuclei segmentation within the domain of histopathological images. The review encompasses a structured analysis of recent literature, focusing on the utilization of YOLO variants for nuclei segmentation. Key methodologies, training strategies, dataset specifics, and performance metrics are evaluated to elucidate the strengths and limitations of YOLO in this context. Additionally, the review highlights the unique characteristics of YOLO that enable efficient object detection and delineation of nuclei structures, offering a comparative analysis against traditional segmentation approaches. This systematic review underscores the promising outcomes achieved through YOLO-based architectures, emphasizing their potential for accurate and rapid nuclei segmentation. Furthermore, it identifies persistent challenges such as handling variances in nuclei appearances, optimizing model architectures for histopathological images, and improving generalization across diverse datasets. Insights derived from this review can provide a foundation for future research directions and enhancements in nuclei segmentation methodologies using YOLO within histopathology, fostering advancements in disease diagnosis and biomedical research.
Collapse
Affiliation(s)
- Shyam Sundar Debsarkar
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, OH, 45229, USA; Department of Computer Science, University of Cincinnati, OH, 45221, USA.
| | - Bruce Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, OH, 45229, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, OH, 45257, USA; Department of Biomedical Informatics, College of Medicine, University of Cincinnati, OH, 45267, USA.
| | - V B Surya Prasath
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, OH, 45229, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, OH, 45257, USA; Department of Biomedical Informatics, College of Medicine, University of Cincinnati, OH, 45267, USA; Department of Computer Science, University of Cincinnati, OH, 45221, USA.
| |
Collapse
|
36
|
Qin X, Liu X, Xiao W, Luo Q, Xia L, Zhang C. Interpretable Deep-learning Model Based on Superb Microvascular Imaging for Noninvasive Diagnosis of Interstitial Fibrosis in Chronic Kidney Disease. Acad Radiol 2025; 32:2730-2738. [PMID: 39690075 DOI: 10.1016/j.acra.2024.11.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/19/2024]
Abstract
RATIONALE AND OBJECTIVES To develop an interpretable deep learning (XDL) model based on superb microvascular imaging (SMI) for the noninvasive diagnosis of the degree of interstitial fibrosis (IF) in chronic kidney disease (CKD). METHODS We included CKD patients who underwent renal biopsy, two-dimensional ultrasound, and SMI examinations between May 2022 and October 2023. Based on the pathological IF score, they were divided into two groups: minimal-mild IF (≤25%) and moderate-severe IF (>25%). An XDL model based on the SMI while establishing an ultrasound radiomics model and a color doppler ultrasonography (CDUS) model as the control group. The utility of the proposed model was evaluated using the receiver operating characteristic curve (ROC) and decision curve analysis. RESULTS In total, 365 CKD patients were included herein. In the validation group, AUCs of the ROC curves for the DL, ultrasound radiomics, and CDUS models were 0.854, 0.784, and 0.745, respectively. In the test group, AUCs of the ROC curve for the DL ultrasound radiomics, and CDUS models were 0.824, 0.792, and 0.752, respectively. The pie chart and heat map based on Shapley additive explanations (SHAP) provided substantial interpretability for the model. CONCLUSION Compared with the ultrasound radiomics and CDUS models, the DL model based on the SMI had higher accuracy in the noninvasive judgment of the degree of IF in CKD patients. Pie and heat maps based on Shapley can explain which image regions are helpful in diagnosing the degree of IF.
Collapse
Affiliation(s)
- Xiachuan Qin
- Department of Ultrasound, Chengdu Second People's Hospital, Chengdu, Sichuan 610000, China (X.Q.); Department of Ultrasound, The first affiliated hospital of Anhui Medical University, Hefei, Anhui 230022, China (X.Q., X.L., Q.L., L.X., C.Z.)
| | - Xiaoling Liu
- Department of Ultrasound, Beijing Anzhen Hospital Nanchong Hospital, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College (University), Nan Chong, Sichuan 637000, China (X.L., W.X.); Department of Ultrasound, The first affiliated hospital of Anhui Medical University, Hefei, Anhui 230022, China (X.Q., X.L., Q.L., L.X., C.Z.)
| | - Weihan Xiao
- Department of Ultrasound, Beijing Anzhen Hospital Nanchong Hospital, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College (University), Nan Chong, Sichuan 637000, China (X.L., W.X.)
| | - Qi Luo
- Department of Ultrasound, The first affiliated hospital of Anhui Medical University, Hefei, Anhui 230022, China (X.Q., X.L., Q.L., L.X., C.Z.)
| | - Linlin Xia
- Department of Ultrasound, The first affiliated hospital of Anhui Medical University, Hefei, Anhui 230022, China (X.Q., X.L., Q.L., L.X., C.Z.)
| | - Chaoxue Zhang
- Department of Ultrasound, The first affiliated hospital of Anhui Medical University, Hefei, Anhui 230022, China (X.Q., X.L., Q.L., L.X., C.Z.).
| |
Collapse
|
37
|
Zhang MX, Liu PF, Zhang MD, Su PG, Shang HS, Zhu JT, Wang DY, Ji XY, Liao QM. Deep learning in nuclear medicine: from imaging to therapy. Ann Nucl Med 2025; 39:424-440. [PMID: 40080372 DOI: 10.1007/s12149-025-02031-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/24/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Deep learning, a leading technology in artificial intelligence (AI), has shown remarkable potential in revolutionizing nuclear medicine. OBJECTIVE This review presents recent advancements in deep learning applications, particularly in nuclear medicine imaging, lesion detection, and radiopharmaceutical therapy. RESULTS Leveraging various neural network architectures, deep learning has significantly enhanced the accuracy of image reconstruction, lesion segmentation, and diagnosis, improving the efficiency of disease detection and treatment planning. The integration of deep learning with functional imaging techniques such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT) enable more precise diagnostics, while facilitating the development of personalized treatment strategies. Despite its promising outlook, there are still some limitations and challenges, particularly in model interpretability, generalization across diverse datasets, multimodal data fusion, and the ethical and legal issues faced in its application. CONCLUSION As technological advancements continue, deep learning is poised to drive substantial changes in nuclear medicine, particularly in the areas of precision healthcare, real-time treatment monitoring, and clinical decision-making. Future research will likely focus on overcoming these challenges and further enhancing model transparency, thus improving clinical applicability.
Collapse
Affiliation(s)
- Meng-Xin Zhang
- Department of Microbiology and Immunology, Henan Provincial Research Center of Engineering Technology for Nuclear Protein Medical Detection, Zhengzhou Health College, Zhengzhou, 45000, Henan, China
- Department of Nuclear Medicine, Henan International Joint Laboratory for Nuclear Protein Regulation, The First Affiliated Hospital, Henan University College of Medicine, Ximen St, Kaifeng, 475004, Henan, China
| | - Peng-Fei Liu
- Department of Microbiology and Immunology, Henan Provincial Research Center of Engineering Technology for Nuclear Protein Medical Detection, Zhengzhou Health College, Zhengzhou, 45000, Henan, China
- Department of Nuclear Medicine, Henan International Joint Laboratory for Nuclear Protein Regulation, The First Affiliated Hospital, Henan University College of Medicine, Ximen St, Kaifeng, 475004, Henan, China
| | - Meng-Di Zhang
- Department of Microbiology and Immunology, Henan Provincial Research Center of Engineering Technology for Nuclear Protein Medical Detection, Zhengzhou Health College, Zhengzhou, 45000, Henan, China
- Department of Nuclear Medicine, Henan International Joint Laboratory for Nuclear Protein Regulation, The First Affiliated Hospital, Henan University College of Medicine, Ximen St, Kaifeng, 475004, Henan, China
| | - Pei-Gen Su
- Department of Microbiology and Immunology, Henan Provincial Research Center of Engineering Technology for Nuclear Protein Medical Detection, Zhengzhou Health College, Zhengzhou, 45000, Henan, China
- School of Medical Technology, Qiqihar Medical University, Qiqihar, 161006, Heilongjiang, China
| | - He-Shan Shang
- Department of Microbiology and Immunology, Henan Provincial Research Center of Engineering Technology for Nuclear Protein Medical Detection, Zhengzhou Health College, Zhengzhou, 45000, Henan, China
- School of Computer and Information Engineering, Henan University, Kaifeng, 475004, Henan, China
| | - Jiang-Tao Zhu
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, 450064, Henan, China.
- Department of Surgery, Faculty of Clinical Medicine, Zhengzhou Shu-Qing Medical College, Gongming Rd, Mazhai Town, Zhengzhou, 450064, Henan, China.
| | - Da-Yong Wang
- Department of Microbiology and Immunology, Henan Provincial Research Center of Engineering Technology for Nuclear Protein Medical Detection, Zhengzhou Health College, Zhengzhou, 45000, Henan, China.
- Department of Nuclear Medicine, Henan International Joint Laboratory for Nuclear Protein Regulation, The First Affiliated Hospital, Henan University College of Medicine, Ximen St, Kaifeng, 475004, Henan, China.
| | - Xin-Ying Ji
- Department of Microbiology and Immunology, Henan Provincial Research Center of Engineering Technology for Nuclear Protein Medical Detection, Zhengzhou Health College, Zhengzhou, 45000, Henan, China.
- Department of Nuclear Medicine, Henan International Joint Laboratory for Nuclear Protein Regulation, The First Affiliated Hospital, Henan University College of Medicine, Ximen St, Kaifeng, 475004, Henan, China.
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, 450064, Henan, China.
| | - Qi-Ming Liao
- Department of Medical Informatics and Computer, Shu-Qing Medical College of Zhengzhou, Gong-Ming Rd, Mazhai Town, Erqi District, Zhengzhou, 450064, Henan, China.
| |
Collapse
|
38
|
Kashiwa W, Hirata K, Endo H, Kudo K, Katoh C, Kawakami T, Kanno H, Takahashi K, Miyazaki T, Ikeda E, Oharaseki T, Ogawa Y, Onimaru M, Kurata M, Nakazawa D, Muso E, Nishibata Y, Masuda S, Tomaru U, Matsuno Y, Furuta S, Abe Y, Tamura N, Harigai M, Ishizu A. Artificial intelligence challenge of discriminating cutaneous arteritis and polyarteritis nodosa based on hematoxylin-and-eosin images of skin biopsy specimens. Pathol Res Pract 2025; 269:155915. [PMID: 40112595 DOI: 10.1016/j.prp.2025.155915] [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: 01/04/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Diseases that develop necrotizing vasculitis of cutaneous muscular arteries include cutaneous arteritis (CA) and polyarteritis nodosa (PAN). It is difficult to distinguish them based on skin biopsy findings alone. This study demonstrated that artificial intelligence (AI) can discriminate them based on skin biopsy findings and revealed where AI focuses on the image. Ninety-three hematoxylin-and-eosin images of CA and 19 PAN images were used. Among them, 85 CA and 17 PAN images were used to train AI; thereafter, AI was challenged to classify the remaining images. The same test images were evaluated by 26 pathologists with different years of experience. AI accuracy was 75.2 %, whereas that of pathologists was 42.8 %. Gradient-weighted class activation mapping (Grad-CAM) indicated that AI focused on connective tissues around the affected vessels rather than the affected vessels. Twenty-two of the 26 pathologists were randomly divided into two groups of 11 each, one of which referred to Grad-CAM images and was challenged in the second-round test of images different from the first round. The accuracy significantly improved after referring to Grad-CAM images, whereas it was equivalent to the first round without referring to Grad-CAM images. In the survey after the second-round test, pathologists who referred to Grad-CAM images suggested that inflammation and fibrosis in the surrounding connective tissues in PAN might be abundant compared to CA. AI may be useful for histological differentiation between CA and PAN and can help pathologists improve the ability of discriminating CA and PAN based on histological findings of skin biopsy specimens.
Collapse
Affiliation(s)
- Wataru Kashiwa
- Deaprtment of Diagnostic Imaging, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kenji Hirata
- Deaprtment of Diagnostic Imaging, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroki Endo
- Deaprtment of Diagnostic Imaging, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kohsuke Kudo
- Deaprtment of Diagnostic Imaging, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Chietsugu Katoh
- Department of Biomedical Science and Engineering, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | | | - Hiroyuki Kanno
- Department of Pathology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kei Takahashi
- Department of Pathology, Toho University Ohashi Medical Center, Tokyo, Japan
| | | | - Eiji Ikeda
- Department of Pathology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Toshiaki Oharaseki
- Department of Pathology, Toho University Ohashi Medical Center, Tokyo, Japan
| | - Yayoi Ogawa
- Hokkaido Renal Pathology Center, Sapporo, Japan
| | - Mitsuho Onimaru
- Division of Pathophysiological and Experimental Pathology, Department of Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mie Kurata
- Department of Analytical Pathology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Daigo Nakazawa
- Department of Rheumatology, Endocrinology, and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Eri Muso
- Department of Nephrology and Dialysis, Medical Research Institute Kitano Hospital, PIIF Tazuke Kofukai, Osaka, Japan
| | - Yuka Nishibata
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Sakiko Masuda
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Utano Tomaru
- Deaprtment of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Yoshihiro Matsuno
- Deaprtment of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Shunsuke Furuta
- Department of Allergy and Clinical Immunology, Chiba University Hospital, Chiba, Japan
| | - Yoshiyuki Abe
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Naoto Tamura
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Masayoshi Harigai
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Akihiro Ishizu
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
39
|
Du S, Liang S, Gu Y. A Language-Guided Progressive Fusion Network with semantic density alignment for Medical Visual Question Answering. J Biomed Inform 2025; 165:104811. [PMID: 40113190 DOI: 10.1016/j.jbi.2025.104811] [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: 11/05/2024] [Revised: 01/17/2025] [Accepted: 03/09/2025] [Indexed: 03/22/2025]
Abstract
Medical Visual Question Answering (Med-VQA) is a critical multimodal task with the potential to address the scarcity and imbalance of medical resources. However, most existing studies overlook the limitations of the inconsistency in information density between medical images and text, as well as the long-tail distribution in datasets, which continue to make Med-VQA an open challenge. To overcome these issues, this study proposes a Language-Guided Progressive Fusion Network (LGPFN) with three key modules: Question-Guided Progressive Multimodal Fusion (QPMF), Language-Gate Mechanism (LGM), and Triple Semantic Feature Alignment (TriSFA). QPMF progressively guides the fusion of visual and textual features using both global and local question representations. LGM, a linguistic rule-based module, distinguishes between Closed-Ended (CE) and Open-Ended (OE) samples, directing the fused features to the appropriate classifiers. Finally, TriSFA captures the rich semantic information of OE answers and mine the underlying associations among fused features, predicted answers, and ground truths, aligning them in a ternary semantic feature space. The proposed LGPFN framework outperforms existing state-of-the-art models, achieving the best overall accuracies of 80.39%, 84.07%, 75.74%, and 70.60% on the VQA-RAD, SLAKE, PathVQA, and VQA-Med 2019 datasets, respectively. These results demonstrate the effectiveness and generalizability of the proposed model, underscoring its potential as a medical Artificial Intelligent (AI) agent that could benefit universal health coverage.
Collapse
Affiliation(s)
- Shuxian Du
- School of Biomedical Engineering, Capital Medical University, Beijing, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing, China; Beijing Key Laboratory of Fundamicationental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing, China.
| | - Shuang Liang
- School of Biomedical Engineering, Capital Medical University, Beijing, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing, China; Beijing Key Laboratory of Fundamicationental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing, China.
| | - Yu Gu
- School of Biomedical Engineering, Capital Medical University, Beijing, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing, China; Beijing Key Laboratory of Fundamicationental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing, China.
| |
Collapse
|
40
|
Anteghini M, Gualdi F, Oliva B. How did we get there? AI applications to biological networks and sequences. Comput Biol Med 2025; 190:110064. [PMID: 40184941 DOI: 10.1016/j.compbiomed.2025.110064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025]
Abstract
The rapidly advancing field of artificial intelligence (AI) has transformed numerous scientific domains, including biology, where a vast and complex volume of data is available for analysis. This paper provides a comprehensive overview of the current state of AI-driven methodologies in genomics, proteomics, and systems biology. We discuss how machine learning algorithms, particularly deep learning models, have enhanced the accuracy and efficiency of embedding sequences, motif discovery, and the prediction of gene expression and protein structure. Additionally, we explore the integration of AI in the embedding and analysis of biological networks, including protein-protein interaction networks and multi-layered networks. By leveraging large-scale biological data, AI techniques have enabled unprecedented insights into complex biological processes and disease mechanisms. This work underlines the potential of applying AI to complex biological data, highlighting current applications and suggesting directions for future research to further explore AI in this rapidly evolving field.
Collapse
Affiliation(s)
- Marco Anteghini
- BioFolD Unit, Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy; Visual and Data-Centric Computing, Zuse Institut Berlin, Berlin, Germany.
| | - Francesco Gualdi
- Structural Bioinformatics Lab, Universitat Pompeu Fabra, Barcelona, Spain; Istituto dalle Molle di Studi sull'Intelligenza Artificiale, USI/SUPSI (Università Svizzera Italiana/Scuola Universitaria Professionale Svizzera Italiana) Lugano, Switzerland.
| | - Baldo Oliva
- Structural Bioinformatics Lab, Universitat Pompeu Fabra, Barcelona, Spain.
| |
Collapse
|
41
|
Sasaki K, Garcia-Manero G, Nigo M, Jabbour E, Ravandi F, Wierda WG, Jain N, Takahashi K, Montalban-Bravo G, Daver NG, Thompson PA, Pemmaraju N, Kontoyiannis DP, Sato J, Karimaghaei S, Soltysiak KA, Raad II, Kantarjian HM, Carter BW. Artificial Intelligence Assessment of Chest Radiographs for COVID-19. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:319-327. [PMID: 39710565 PMCID: PMC11993350 DOI: 10.1016/j.clml.2024.11.013] [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: 01/24/2022] [Revised: 10/21/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND The sensitivity of reverse-transcription polymerase chain reaction (RT-PCR) is limited for diagnosis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Chest computed tomography (CT) is reported to have high sensitivity; however, given the limited availability of chest CT during a pandemic, the assessment of more readily available imaging, such as chest radiographs, augmented by artificial intelligence may substitute for the detection of the features of coronavirus disease 2019 (COVID-19) pneumonia. METHODS We trained a deep convolutional neural network to detect SARS-CoV-2 pneumonia using publicly available chest radiography imaging data including 8,851 normal, 6,045 pneumonia, and 200 COVID-19 pneumonia radiographs. The entire cohort was divided into training (n = 13,586) and test groups (n = 1510). We assessed the accuracy of prediction with independent external data. RESULTS The sensitivity and positive predictive values of the assessment by artificial intelligence were 96.8% and 90.9%, respectively. In the first external validation of 204 chest radiographs among 107 patients with confirmed COVID-19, the artificial intelligence algorithm correctly identified 174 (85%) chest radiographs as COVID-19 pneumonia among 97 (91%) patients. In the second external validation with 50 immunocompromised patients with leukemia, the higher probability of the artificial intelligence assessment for COVID-19 was correlated with suggestive features of COVID-19, while the normal chest radiographs were closely correlated with the likelihood of normal chest radiographs by the artificial intelligence prediction. CONCLUSIONS The assessment method by artificial intelligence identified suspicious lung lesions on chest radiographs. This novel approach can identify patients for confirmatory chest CT before the progression of COVID-19 pneumonia.
Collapse
Affiliation(s)
- Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| | | | - Masayuki Nigo
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - William G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Philip A Thompson
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dimitrios P Kontoyiannis
- Department of Infectious Disease, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Junya Sato
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sam Karimaghaei
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX
| | - Kelly A Soltysiak
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Issam I Raad
- Department of Infectious Disease, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Brett W Carter
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
42
|
He R, Cao J, Tan T. Generative artificial intelligence: a historical perspective. Natl Sci Rev 2025; 12:nwaf050. [PMID: 40191253 PMCID: PMC11970245 DOI: 10.1093/nsr/nwaf050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/05/2025] [Accepted: 02/09/2025] [Indexed: 04/09/2025] Open
Abstract
Generative artificial intelligence (GAI) has recently achieved significant success, enabling anyone to create texts, images, videos and even computer codes while providing insights that might not be possible with traditional tools. To stimulate future research, this work provides a brief summary of the ongoing and historical developments in GAI over the past 70 years. The achievements are grouped into four categories: (i) rule-based generative systems that follow specialized rules and instructions, (ii) model-based generative algorithms that produce new content based on statistical or graphical models, (iii) deep generative methodologies that utilize deep neural networks to learn how to generate new content from data and (iv) foundation models that are trained on extensive datasets and capable of performing a variety of generative tasks. This paper also reviews successful generative applications and identifies open challenges posed by remaining issues. In addition, this paper describes potential research directions aimed at better utilizing, understanding and harnessing GAI technologies.
Collapse
Affiliation(s)
- Ran He
- New Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Intelligence Science and Technology, Nanjing University, Nanjing 210008, China
| | - Jie Cao
- New Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Tieniu Tan
- New Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Intelligence Science and Technology, Nanjing University, Nanjing 210008, China
| |
Collapse
|
43
|
Ver Berne J, Saadi SB, Oliveira-Santos N, Marinho-Vieira LE, Jacobs R. Automated classification of panoramic radiographs with inflammatory periapical lesions using a CNN-LSTM architecture. J Dent 2025; 156:105688. [PMID: 40101853 DOI: 10.1016/j.jdent.2025.105688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
OBJECTIVES Considering Convolutional Neural Network (CNN) and Long Short-Term Memory (LSTM) network approaches have shown promising image classification performance, the aim of this study was to compare the performance of novel Convolutional Neural Network and Long Short-Term Memory (CNN-LSTM) architectures with a classic CNN for classification of panoramic radiographs with inflammatory periapical lesions. METHODS A dataset of 356 panoramic radiographs with periapical lesions and 769 control images were retrospectively collected and divided into training, validation, and testing sets. Next, four different models were constructed: a classic CNN, a classic LSTM, a cascaded CNN-LSTM, and parallel CNN-LSTM architecture. In each model the CNN took the full panoramic radiograph as input while the LSTM network ran on the images divided into 6 sequential patches. Sensitivity, specificity, and Area Under the Receiver-Operating Curve (AUC) were calculated. McNemar's test compared the sensitivity and specificity between the classic CNN and the other models. RESULTS Parallel CNN-LSTM had a significantly higher sensitivity than classic CNN for detecting periapical lesions (95% vs. 81%, 95% confidence interval for the difference = 6 - 22 %, P = 0.002), while also exhibiting the best overall performance of the four models [AUC = 96% vs. 90% (classic CNN), 92% (classic LSTM), and 94% (cascaded CNN-LSTM)]. CONCLUSIONS The parallel CNN-LSTM architecture outperformed the classic CNN for classification of panoramic radiographs with inflammatory periapical lesions. CLINICAL SIGNIFICANCE Combining CNN and LSTM models improves the classification of panoramic radiographs with and without inflammatory periapical lesions.
Collapse
Affiliation(s)
- Jonas Ver Berne
- OMFS-IMPATH Research Group, Department of Imaging & Pathology, Catholic University Leuven, Belgium; Department of Oral & Maxillofacial Surgery, University Hospitals Leuven, Belgium
| | - Soroush Baseri Saadi
- Department of Oral & Maxillofacial Surgery, University Hospitals Leuven, Belgium
| | - Nicolly Oliveira-Santos
- OMFS-IMPATH Research Group, Department of Imaging & Pathology, Catholic University Leuven, Belgium; Department of Oral Surgery & Stomatology, Division of Oral Diagnostic Sciences, School of Dental Medicine, University of Bern, Switzerland
| | - Luiz Eduardo Marinho-Vieira
- OMFS-IMPATH Research Group, Department of Imaging & Pathology, Catholic University Leuven, Belgium; Division of Oral Radiology, Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Brazil
| | - Reinhilde Jacobs
- OMFS-IMPATH Research Group, Department of Imaging & Pathology, Catholic University Leuven, Belgium; Department of Oral & Maxillofacial Surgery, University Hospitals Leuven, Belgium; Department of Dental Medicine, Karolinska Institutet, Sweden.
| |
Collapse
|
44
|
Bauer F, Kächele J, Bernhard J, Hajiyianni M, Weinhold N, Sauer S, Grözinger M, Raab MS, Mai EK, Weber TF, Goldschmidt H, Schlemmer HP, Maier-Hein K, Delorme S, Neher P, Wennmann M. Advanced Automated Model for Robust Bone Marrow Segmentation in Whole-body MRI. Acad Radiol 2025; 32:2824-2835. [PMID: 39848889 DOI: 10.1016/j.acra.2024.12.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/19/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025]
Abstract
RATIONALE AND OBJECTIVES To establish an advanced automated bone marrow (BM) segmentation model on whole-body (WB-)MRI in monoclonal plasma cell disorders (MPCD), and to demonstrate its robust performance on multicenter datasets with severe myeloma-related pathologies. MATERIALS AND METHODS The study cohort comprised multi-vendor, multi-protocol imaging data acquired with varying field strength across 8 different centers. In total, 210 WB-MRIs of 207 MPCD patients were included. An nnU-Net algorithm was established for segmenting the individual bone marrow spaces (BMS) of the spine, pelvis, humeri and femora (advanced segmentation model). For this task, 186 T1-weighted (T1w) WB-MRIs from center 1 were used in the training set. Test sets included 12 T1w WB-MRIs from center 2 (I) and 9 T1w WB-MRIs from centers 3-8 (II). Example cases were included to showcase segmentation performance on T1w WB-MRIs with extensive tumor load. The segmentation accuracy of the advanced segmentation model was compared to a prior established basic segmentation model by calculating Dice scores and using the Wilcoxon signed-rank test. RESULTS The mean Dice score on the individual BMS was 0.89±0.13 (test set I) and 0.88±0.11 (test set II), significantly higher than the Dice scores of a prior basic model (p<0.05). Dice scores for the BMS of the individual bones ranged from 0.77 to 0.96 (test set I), and 0.81 to 0.95 (test set II). BM altered by myeloma-relevant pathologies, artifacts or low imaging quality was precisely segmented. CONCLUSION The advanced model performed reliable, automated segmentations, even on heterogeneously acquired multicenter WB-MRIs with severe pathologies.
Collapse
Affiliation(s)
- Fabian Bauer
- Division of Radiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany (F.B., M.G., H.P.S., S.D.); Division of Musculoskeletal Imaging and Intervention, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 (F.B.).
| | - Jessica Kächele
- Division of Medical Image Computing, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany (J.K., J.B., K.M.H., P.N.); German Cancer Consortium (DKTK), Partner Site Heidelberg, 69120 Heidelberg, Germany (J.K., K.M.H., P.N.)
| | - Juliane Bernhard
- Division of Medical Image Computing, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany (J.K., J.B., K.M.H., P.N.)
| | - Marina Hajiyianni
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.H., N.W., S.S., M.S.R., E.K.M., H.G.)
| | - Niels Weinhold
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.H., N.W., S.S., M.S.R., E.K.M., H.G.)
| | - Sandra Sauer
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.H., N.W., S.S., M.S.R., E.K.M., H.G.)
| | - Martin Grözinger
- Division of Radiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany (F.B., M.G., H.P.S., S.D.)
| | - Marc-Steffen Raab
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.H., N.W., S.S., M.S.R., E.K.M., H.G.)
| | - Elias K Mai
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.H., N.W., S.S., M.S.R., E.K.M., H.G.)
| | - Tim F Weber
- Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany (T.F.W., M.W.)
| | - Hartmut Goldschmidt
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.H., N.W., S.S., M.S.R., E.K.M., H.G.); National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120 Heidelberg, Germany (H.G., K.M.H., P.N.)
| | - Heinz-Peter Schlemmer
- Division of Radiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany (F.B., M.G., H.P.S., S.D.)
| | - Klaus Maier-Hein
- Division of Medical Image Computing, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany (J.K., J.B., K.M.H., P.N.); German Cancer Consortium (DKTK), Partner Site Heidelberg, 69120 Heidelberg, Germany (J.K., K.M.H., P.N.); National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120 Heidelberg, Germany (H.G., K.M.H., P.N.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany (K.M.H., P.N.)
| | - Stefan Delorme
- Division of Radiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany (F.B., M.G., H.P.S., S.D.)
| | - Peter Neher
- Division of Medical Image Computing, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany (J.K., J.B., K.M.H., P.N.); German Cancer Consortium (DKTK), Partner Site Heidelberg, 69120 Heidelberg, Germany (J.K., K.M.H., P.N.); National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120 Heidelberg, Germany (H.G., K.M.H., P.N.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany (K.M.H., P.N.)
| | - Markus Wennmann
- Division of Radiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany (F.B., M.G., H.P.S., S.D.); Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany (T.F.W., M.W.)
| |
Collapse
|
45
|
Godwin RC, Tung A, Berkowitz DE, Melvin RL. Transforming Physiology and Healthcare through Foundation Models. Physiology (Bethesda) 2025; 40:0. [PMID: 39832521 DOI: 10.1152/physiol.00048.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/30/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
Recent developments in artificial intelligence (AI) may significantly alter physiological research and healthcare delivery. Whereas AI applications in medicine have historically been trained for specific tasks, recent technological advances have produced models trained on more diverse datasets with much higher parameter counts. These new, "foundation" models raise the possibility that more flexible AI tools can be applied to a wider set of healthcare tasks than in the past. This review describes how these newer models differ from conventional task-specific AI, which relies heavily on focused datasets and narrow, specific applications. By examining the integration of AI into diagnostic tools, personalized treatment strategies, biomedical research, and healthcare administration, we highlight how these newer models are revolutionizing predictive healthcare analytics and operational workflows. In addition, we address ethical and practical considerations associated with the use of foundation models by highlighting emerging trends, calling for changes to existing guidelines, and emphasizing the importance of aligning AI with clinical goals to ensure its responsible and effective use.
Collapse
Affiliation(s)
- Ryan C Godwin
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Avery Tung
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, Illinois, United States
| | - Dan E Berkowitz
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Ryan L Melvin
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
46
|
Genc A, Marlowe J, Jalil A, Belzberg D, Kovarik L, Christopher P. A versatile machine learning workflow for high-throughput analysis of supported metal catalyst particles. Ultramicroscopy 2025; 271:114116. [PMID: 40014985 DOI: 10.1016/j.ultramic.2025.114116] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/29/2025] [Accepted: 02/15/2025] [Indexed: 03/01/2025]
Abstract
Accurate and efficient characterization of nanoparticles (NPs), particularly regarding particle size distribution, is essential for advancing our understanding of their structure-property relationship and facilitating their design for various applications. In this study, we introduce a novel two-stage artificial intelligence (AI)-driven workflow for NP analysis that leverages prompt engineering techniques from state-of-the-art single-stage object detection and large-scale vision transformer (ViT) architectures. This methodology is applied to transmission electron microscopy (TEM) and scanning TEM (STEM) images of heterogeneous catalysts, enabling high-resolution, high-throughput analysis of particle size distributions for supported metal catalyst NPs. The model's performance in detecting and segmenting NPs is validated across diverse heterogeneous catalyst systems, including various metals (Ru, Cu, PtCo, and Pt), supports (silica (SiO2), γ-alumina (γ-Al2O3), and carbon black), and particle diameter size distributions with mean and standard deviations ranging from 1.6 ± 0.2 nm to 9.7 ± 4.6 nm. The proposed machine learning (ML) methodology achieved an average F1 overlap score of 0.91 ± 0.01 and demonstrated the ability to disentangle overlapping NPs anchored on catalytic support materials. The segmentation accuracy is further validated using the Hausdorff distance and robust Hausdorff distance metrics, with the 90th percent of the robust Hausdorff distance showing errors within 0.4 ± 0.1 nm to 1.4 ± 0.6 nm. Our AI-assisted NP analysis workflow demonstrates robust generalization across diverse datasets and can be readily applied to similar NP segmentation tasks without requiring costly model retraining.
Collapse
Affiliation(s)
- Arda Genc
- Materials Department, University of California, Santa Barbara, CA, USA.
| | - Justin Marlowe
- Department of Chemical Engineering, University of California, Santa Barbara, CA, USA
| | - Anika Jalil
- Department of Chemical Engineering, University of California, Santa Barbara, CA, USA
| | - Daniel Belzberg
- Department of Physics, University of California, Santa Barbara, CA, USA
| | - Libor Kovarik
- Institute for Integrated Catalysis, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Phillip Christopher
- Department of Chemical Engineering, University of California, Santa Barbara, CA, USA
| |
Collapse
|
47
|
Jiang Y, Sun HT, Luo Z, Wang J, Xiao RP. Efficacy of a deep learning system for automatic analysis of the comprehensive spatial relationship between the mandibular third molar and inferior alveolar canal on panoramic radiographs. Oral Surg Oral Med Oral Pathol Oral Radiol 2025; 139:612-622. [PMID: 39915134 DOI: 10.1016/j.oooo.2024.12.020] [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: 09/02/2024] [Revised: 12/10/2024] [Accepted: 12/24/2024] [Indexed: 03/18/2025]
Abstract
OBJECTIVE To develop and evaluate a deep learning (DL) system for predicting the contact and relative position relationships between the mandibular third molar (M3) and inferior alveolar canal (IAC) using panoramic radiographs (PRs) for preoperative assessment of patients for M3 surgery. STUDY DESIGN In total, 279 PRs with 441 M3s from individuals aged 18-32 years were collected, with one PR and cone beam computed tomography (CBCT) scan per individual. Six DL models were compared using 5-fold cross-validation. Model performance was evaluated using accuracy, precision, recall, specificity, F1-score, and area under the receiver operating characteristic (AUROC) curve. System performance was compared to that of experienced dentists. The diagnostic performance was investigated based on the reference standard for contact and relative position between M3 and IAC as determined by CBCT. RESULTS ResNet50 exhibited the best performance among all models tested. For contact prediction, ResNet50 achieved an accuracy of 0.748, F1-score of 0.759, and AUROC of 0.811. For relative position relationship prediction, ResNet50 yielded an accuracy of 0.611, F1-score of 0.548, and AUROC of 0.731. The DL system demonstrated advantages over experienced dentists in diagnostic outcomes. CONCLUSIONS The developed DL system shows broad application potential for comprehensive spatial relationship recognition between M3 and IAC. This system can assist dentists in treatment decision-making for M3 surgery and improve dentist training efficiency.
Collapse
Affiliation(s)
- Yi Jiang
- College of Future Technology, Peking University, Beijing, China
| | - Hai-Tao Sun
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Zhengchao Luo
- College of Future Technology, Peking University, Beijing, China
| | - Jinzhuo Wang
- College of Future Technology, Peking University, Beijing, China.
| | - Rui-Ping Xiao
- College of Future Technology, Peking University, Beijing, China.
| |
Collapse
|
48
|
Girmatsion M, Tang X, Zhang Q, Li P. Progress in machine learning-supported electronic nose and hyperspectral imaging technologies for food safety assessment: A review. Food Res Int 2025; 209:116285. [PMID: 40253192 DOI: 10.1016/j.foodres.2025.116285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/08/2025] [Accepted: 03/12/2025] [Indexed: 04/21/2025]
Abstract
The growing concern over food safety, driven by threats such as food contaminations and adulterations has prompted the adoption of advanced technologies like electronic nose (e-nose) and hyperspectral imaging (HSI), which are increasingly enhanced by machine learning innovations. This paper aims to provide a comprehensive review on food safety, by combining insights from both e-nose and HSI technologies alongside machine learning algorithms. First, the basic principles of e-nose, HSI, and machine learning, with particular emphasis on artificial neural network (ANN) and deep learning (DL) are briefly discussed. The review then examines how machine learning enhances the performance of e-nose and HSI, followed by an exploration of recent applications in detecting food hazards, including drug residues, microbial contaminants, pesticide residues, toxins, and adulterants. Subsequently, key limitations encountered in the applications of machine learning, e-nose and HSI, along with future perspectives on the potential advancements of these technologies are highlighted. E-nose and HSI technologies have shown their great potential for applications in food safety assessment through machine learning assistance. Despite this, their use is primarily limited to laboratory environments, restricting their real-world applications. Additionally, the lack of standardized protocols hampers their acceptance and the reproducibility of tests in food safety assessments. Thus, further research is essential to address these limitations and enhance the effectiveness of e-nose and HSI technologies in practical applications. Ultimately, this paper offers a detailed understanding of both technologies, highlighting the pivotal role of machine learning and presenting insights into their innovative applications within food safety evaluation.
Collapse
Affiliation(s)
- Mogos Girmatsion
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China; Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture and Rural Affairs; Laboratory of Risk Assessment for Oilseed Products (Wuhan), Ministry of Agriculture and Rural Affairs; Quality Inspection and Test Center for Oilseed Products, Ministry of Agriculture and Rural Affairs, China; Hamelmalo Agricultural College, Department of Food Science, Keren, Eritrea
| | - Xiaoqian Tang
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China; Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture and Rural Affairs; Laboratory of Risk Assessment for Oilseed Products (Wuhan), Ministry of Agriculture and Rural Affairs; Quality Inspection and Test Center for Oilseed Products, Ministry of Agriculture and Rural Affairs, China; Food Safety Research Institute, Hubei University, Wuhan 430062, China; Hubei Hongshan Laboratory, Wuhan 430070, China.
| | - Qi Zhang
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China; Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture and Rural Affairs; Laboratory of Risk Assessment for Oilseed Products (Wuhan), Ministry of Agriculture and Rural Affairs; Quality Inspection and Test Center for Oilseed Products, Ministry of Agriculture and Rural Affairs, China; Food Safety Research Institute, Hubei University, Wuhan 430062, China; Hubei Hongshan Laboratory, Wuhan 430070, China; Xianghu Laboratory, Hangzhou 311231, China
| | - Peiwu Li
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China; Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture and Rural Affairs; Laboratory of Risk Assessment for Oilseed Products (Wuhan), Ministry of Agriculture and Rural Affairs; Quality Inspection and Test Center for Oilseed Products, Ministry of Agriculture and Rural Affairs, China; Food Safety Research Institute, Hubei University, Wuhan 430062, China; Hubei Hongshan Laboratory, Wuhan 430070, China; Xianghu Laboratory, Hangzhou 311231, China.
| |
Collapse
|
49
|
Rahmatullah GM, Ruan SJ, Wisesa IWW, Li LPH. Enhancing visual speech perception through deep automatic lipreading: A systematic review. Comput Biol Med 2025; 190:110019. [PMID: 40157316 DOI: 10.1016/j.compbiomed.2025.110019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 01/24/2025] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
Communication involves exchanging information between individuals or groups through various media sources. However, limitations such as hearing loss can make it difficult for some individuals to understand the information delivered during speech communication. Conventional methods, including sign language, written text, and manual lipreading, offer some solutions; however, emerging software-based tools using artificial intelligence (AI) are introducing more effective approaches. Many approaches rely on AI to improve communication quality, with the current trend of leveraging deep learning being a particularly effective tool. This paper presents a comprehensive Systematic Literature Review (SLR) of research trends in automatic lipreading technologies, a critical field in enhancing communication among individuals with hearing impairments. The SLR, which followed the Preferred Reporting Items for Systematic Literature Review and Meta-Analysis (PRISMA) protocol, identified 114 original research articles published between 2014 and mid-2024. The essential information from these articles was summarized, including the trends in automatic lipreading research, dataset availability, task categories, existing approaches, and architectures for automatic lipreading systems. The results showed that various techniques and advanced deep learning models achieved convincing performance to become state-of-the-art in automatic lipreading tasks. However, several challenges, such as insufficient data quantity, inadequate environmental conditions, and language diversity, must be resolved in the future. Furthermore, many improvements have been made to the deep learning models to overcome these challenges and become a massive solution, particularly for automatic lipreading tasks in the near future.
Collapse
Affiliation(s)
- Griffani Megiyanto Rahmatullah
- Department of Electronic and Computer Engineering, National Taiwan University of Science and Technology, No. 43, Sec. 4, Keelung Rd, Taipei, 10607, Taiwan; Electrical Electronic Engineering, Politeknik Negeri Bandung, Jl. Gegerkalong Hilir, Ciwaruga, Kec. Parongpong, Bandung, 40012, West Java, Indonesia
| | - Shanq-Jang Ruan
- Department of Electronic and Computer Engineering, National Taiwan University of Science and Technology, No. 43, Sec. 4, Keelung Rd, Taipei, 10607, Taiwan
| | - I Wayan Wiprayoga Wisesa
- Department of Electronic and Computer Engineering, National Taiwan University of Science and Technology, No. 43, Sec. 4, Keelung Rd, Taipei, 10607, Taiwan; Department of Informatics, Institut Teknologi Sumatera, Jl. Terusan Ryacudu, Way Huwi, Kec. Jati Agung, Lampung Selatan, 35365, Lampung, Indonesia
| | - Lieber Po-Hung Li
- Faculty of Medicine and Institute of Brain Science, National Yang-Ming Chiao-Tung University, No. 155, Sec. 2, Linong St, Beitou District, Taipei, 112, Taiwan; Department of Otolaryngology, Cheng Hsin General Hospital, No. 45, Zhenxing St, Beitou District, Taipei, 112, Taiwan; Department of Medical Research, China Medical University Hospital, No. 2, Yude Rd., North District, Taichung, 404327, Taiwan.
| |
Collapse
|
50
|
Mao L. Informing Deep Learning of Sensing Data with Physics and Chemistry. ACS Sens 2025; 10:2386-2387. [PMID: 40275811 DOI: 10.1021/acssensors.5c01075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
|