1
|
Xu C, Xu X, Huang Y, Shang S, Ma L. RNA methylation: A new promising biomaker in cancer liquid biopsy. Biochim Biophys Acta Rev Cancer 2025; 1880:189337. [PMID: 40315965 DOI: 10.1016/j.bbcan.2025.189337] [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: 01/24/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/04/2025]
Abstract
RNA methylation is a vital epigenetic modification that regulates gene expression by influencing RNA processes such as transcription, degradation, translation, and transport. Aberrant methylation, including modifications like m6A, m5C, m1A, m7G, and m3C, is closely linked to tumorigenesis and progression. Liquid biopsy, a non-invasive technique analyzing tumor markers in body fluids, offers significant potential for early diagnosis and dynamic monitoring. In this context, RNA methylation, due to its tumor-specific properties, is emerging as a valuable marker. However, significant challenges remain in its clinical application. This review explores the roles of RNA methylation in cancer, recent advances in detection technologies, and its potential as a liquid biopsy marker in tumor management. It highlights its promising applications in cancer diagnosis, prognosis, and personalized treatment in the era of precision oncology.
Collapse
Affiliation(s)
- Chenxin Xu
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xin Xu
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yiwen Huang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Shuang Shang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Lifang Ma
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
2
|
Shen K, Hu C, Zhang Y, Cheng X, Xu Z, Pan S. Advances and applications of multiomics technologies in precision diagnosis and treatment for gastric cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189336. [PMID: 40311712 DOI: 10.1016/j.bbcan.2025.189336] [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: 01/25/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025]
Abstract
Gastric cancer (GC), one of the most prevalent malignancies worldwide, is distinguished by extensive genetic and phenotypic heterogeneity, posing persistent challenges to conventional diagnostic and therapeutic strategies. The significant global burden of GC highlights an urgent need to unravel its complex underlying mechanisms, discover novel diagnostic and prognostic biomarkers, and develop more effective therapeutic interventions. In this context, this review comprehensively examines the transformative roles of cutting-edge technologies, including radiomics, pathomics, genomics, transcriptomics, epigenomics, proteomics, and metabolomics, in advancing precision diagnosis and treatment for GC. Multiomics data analysis not only deepens our understanding of GC pathogenesis and molecular subtypes but also identifies promising biomarkers, facilitating the creation of tailored therapeutic approaches. Additionally, integrating multiomics approaches holds immense potential for elucidating drug resistance mechanisms, predicting patient outcomes, and uncovering novel therapeutic targets, thereby laying a robust foundation for precision medicine in the comprehensive management of GC.
Collapse
Affiliation(s)
- Ke Shen
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
| | - Can Hu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
| | - Yanqiang Zhang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
| | - Zhiyuan Xu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Siwei Pan
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
3
|
Bhutani K, Vishwakarma S, Yadav P, Yadav MK. The current landscape of aromatase inhibitors for the treatment of estrogen receptor-positive breast carcinoma. J Steroid Biochem Mol Biol 2025; 250:106729. [PMID: 40056742 DOI: 10.1016/j.jsbmb.2025.106729] [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: 01/10/2025] [Revised: 02/18/2025] [Accepted: 03/06/2025] [Indexed: 03/10/2025]
Abstract
Estrogen receptor-positive (ER+) breast carcinoma represents a significant portion of breast cancer cases and is characterized by the presence of estrogen receptors that promote tumor growth upon estrogen binding. ER + breast cancer progression involves hormonal influences, interactions within the tumor microenvironment, and genetic mutations that may lead to treatment resistance. Successful therapeutic options include hormonal therapies, particularly aromatase inhibitors (AIs), which aim to block the effects of estrogen or reduce its synthesis. With higher efficacy than tamoxifen, AIs such as anastrozole, letrozole, and exemestane have become widely employed in adjuvant and first-line treatments for advanced breast cancer. AIs function by inhibiting the enzyme aromatase, which converts androgens into estrogens in the peripheral tissues. Because too much estrogen might promote tumor growth, this decrease in estrogen levels is essential for treating ER+ malignancies. To provide a comprehensive overview of AIs in the treatment of ER+ breast cancer, this study examined the pharmacokinetics, clinical uses, mechanisms of action, and problems with treatment resistance. To maximize therapeutic approaches and enhance patient outcomes in the treatment of ER breast cancer, it is imperative to understand these characteristics.
Collapse
Affiliation(s)
- Khushboo Bhutani
- Department of Biotechnology, SRM University, Delhi-NCR, Sonepat, Haryana 131029, India
| | - Suyashi Vishwakarma
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh 201309, India
| | - Priyanka Yadav
- Department of Biotechnology, SRM University, Delhi-NCR, Sonepat, Haryana 131029, India
| | - Manoj Kumar Yadav
- Department of Biotechnology, SRM University, Delhi-NCR, Sonepat, Haryana 131029, India; Department of Biomedical Engineering, SRM University, Sonepat, Haryana 131029, India.
| |
Collapse
|
4
|
Lu X, Pan C, Yao L, Wan J, Xu X, Wang W, Wang X, Liu X, Jin Z, Wang H, He Y, Yang B. Integrating multimodal data to predict the progression of hormone-sensitive prostate cancer. Clin Proteomics 2025; 22:21. [PMID: 40442579 PMCID: PMC12121097 DOI: 10.1186/s12014-025-09543-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 05/11/2025] [Indexed: 06/02/2025] Open
Abstract
Identifying the population at risk of rapid progression from hormone-sensitive prostate cancer (HSPC) to lethal castration-resistant prostate cancer (CRPC) is a challenge. This work has highlighted important prognostic insights based on proteomics data, magnetic resonance imaging (MRI) and histopathological specimens. We retrospectively developed a multi-omics-based model based on 77 patients with HSPC. In order to identify the features related to survival time under each mode, we used the Boruta algorithm for feature screening. In order to demonstrate the effectiveness of our selected features, we used six machine learning methods to validate the classification of the selected features for each mode. A total of 63 proteome signatures, 60 HE signatures, 56 T2WI signatures, and 54 ADC signatures were identified as features related to the speed of HSPC progression. Ultimately, 30 multi-omics-based features were determined by the least absolute shrinkage and selection operator (LASSO) method and multivariate cox regression. In order to stratify patients with significant disparities in progress, a nomogram model was developed, of which the C-index was 0.906. Accordingly, the developed model could help identify patients who are at a high risk of rapid CRPC progression, and aid clinicians in guiding personalized clinical management and decision-making.
Collapse
Affiliation(s)
- Xiangfu Lu
- Department of Urology, 967 th hospital of PLA Joint Logistics Support Force, No.80 Shengli Road, Dalian, 116014, PR China
| | - Chenxi Pan
- State key laboratory of fine chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, PR China
| | - Luhan Yao
- School of Information and Communication Engineering, Dalian University of Technology, Dalian, Dalian, 116023, PR China
| | - Jiayu Wan
- School of Information and Communication Engineering, Dalian University of Technology, Dalian, Dalian, 116023, PR China
| | - Xiaolong Xu
- Department of Urology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China
| | - Wei Wang
- Department of Urology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China
| | - Xiangying Wang
- State key laboratory of fine chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, PR China
| | - Xiaoyun Liu
- Department of Urology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China
| | - Zhonghua Jin
- Department of chest surgery, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China
| | - Hongyu Wang
- School of Information and Communication Engineering, Dalian University of Technology, Dalian, Dalian, 116023, PR China.
| | - Yi He
- Department of Urology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China.
- The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116023, Liaoning, China.
| | - Bo Yang
- Department of Urology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China.
- The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116023, Liaoning, China.
| |
Collapse
|
5
|
Nopour R. Prediction of one-year recurrence among breast cancer patients undergone surgery using artificial intelligence-based algorithms: a retrospective study on prognostic factors. BMC Cancer 2025; 25:940. [PMID: 40419997 PMCID: PMC12105147 DOI: 10.1186/s12885-025-14369-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 05/20/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND AND AIM Breast cancer is highly prevalent, with an increasing trend in women globally. Although the survival of breast cancer is relatively high, the recurrence rate is also high, demanding effective predictive solutions to breast cancer prognosis among post-operative patients. So far, Artificial intelligence algorithms integrated with various clinical data have demonstrated potential predictive capability regarding breast cancer recurrence. OBJECTIVE This study aims to specifically conduct a predictive analysis of one-year recurrence of breast cancer by comparing and analyzing different machine learning and deep learning algorithms trained by structural prognostic data. MATERIALS AND METHODS This retrospective study was carried out using one database, including 1156 post-operative breast cancer data from 30 January 2020 to 30 December 2022, in three clinical centers in Tehran City. The inclusion criteria were patients who had undergone at least one surgery, had at least one year of medical records, and did not have other conditions. The patients who were diagnosed with malignant BC and had undergone adjuvant therapies without surgery were excluded from the study. Twenty-three prognostic factors were utilized to train algorithms to establish prediction models for the one-year recurrence of breast cancer. The data were analyzed using univariate and adjusted correlation-based methods and chosen machine learning and deep learning algorithms. The discrimination, calibration, and clinical utility were leveraged to assess the algorithms' performance efficiency. The SHapley Additive exPlanations plot was generated to identify the prominent prognostic factors affecting the one-year recurrence of breast cancer. RESULTS Totally, 445 relapsed and 711 non-relapsed cases were utilized in this study. Our empirical study showed that the random forest with a positive predictive value of 0.96, negative predictive value of 0.92, sensitivity of 0.92, specificity of 0.96, accuracy of 0.94, F-score of 0.94, area under the receiver operator characteristics curve of 0.919 was the best-performing model for predicting the breast cancer recurrence. As the analysis of SHapley Additive exPlanations indicated, the tumor grade, HER-2, and the number of lymph nodes involved were more significant predictors. CONCLUSION The current study demonstrated the potential predictive power of the random forest for early predicting tumors among breast cancer patients who have undergone surgery and its utility in enhancing decision-making in clinical environments. It is crucial in promoting the prognosis, more effectively choosing therapies, augmenting post-operative breast cancer patients' survival, and controlling the limited healthcare resources. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Raoof Nopour
- Department of Health Information Management, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Alaimo A, Carbone FG, Buzo K, Annesi N, Genovesi S, Lorenzato A, Widmann K, Libergoli M, Marmocchi E, Bertalot G, Brolese A, Papotti MG, Molinaro L, Caffo O, Barbareschi M, Bardelli A, Romanel A, Arena S, Lunardi A. TRPM8 levels determine tumor vulnerability to channel agonists. Mol Oncol 2025. [PMID: 40405392 DOI: 10.1002/1878-0261.70049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 02/10/2025] [Accepted: 03/30/2025] [Indexed: 05/24/2025] Open
Abstract
Targeted therapies have pervasively enhanced clinical protocols and significantly improved survival and quality of life of cancer patients. Mostly grounded on small molecules and antibodies targeting deregulated mechanisms in cancer cells, precision oncology approaches are limited to a few tumor types because of the paucity of clinically actionable targets. Here, we report a comparative analysis of the cation channel transient receptor potential melastatin 8 (TRPM8; also known as transient receptor potential cation channel subfamily M member 8) in lung, breast, colorectal, and prostate cancers. Our findings reveal high levels of channel expression in cores of all four carcinomas, irrespective of reduced expression of its RNA. Importantly, cancer cell lines that represent the various tumor types consistently show that sub-lethal chemotherapy dosages combined with the TRPM8 agonist D-3263 have a synergistic lethal effect. In addition, administration of D-3263 increases the cytotoxicity of 5-FU/Oxaliplatin in patient-derived colorectal cancer organoids, depending on the levels of TRPM8. Overall, our study strengthens the candidacy of TRPM8 as a molecular target for precision oncology approaches and paves the way for the design of basket trials for its clinical testing in TRPM8-high tumors.
Collapse
Affiliation(s)
- Alessandro Alaimo
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Italy
| | | | - Kristi Buzo
- Department of Oncology, University of Torino, Torino, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Nicole Annesi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Italy
| | - Sacha Genovesi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Italy
| | | | - Karen Widmann
- Surgical Pathology, Santa Chiara Hospital-APSS, Trento, Italy
| | - Michela Libergoli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Italy
| | - Elisa Marmocchi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Italy
| | - Giovanni Bertalot
- Surgical Pathology, Santa Chiara Hospital-APSS, Trento, Italy
- Centre for Medical Sciences-CISMed, University of Trento, Italy
| | - Alberto Brolese
- Department of General Surgery & HPB Unit, Santa Chiara Hospital-APSS, Trento, Italy
| | - Mauro Giulio Papotti
- Department of Pathology, University of Torino and AOU Città della Salute e della Scienza di Torino, Italy
| | - Luca Molinaro
- Department of Pathology, University of Torino and AOU Città della Salute e della Scienza di Torino, Italy
| | - Orazio Caffo
- Medical Oncology, Santa Chiara Hospital-APSS, Trento, Italy
| | - Mattia Barbareschi
- Surgical Pathology, Santa Chiara Hospital-APSS, Trento, Italy
- Centre for Medical Sciences-CISMed, University of Trento, Italy
| | - Alberto Bardelli
- Department of Oncology, University of Torino, Torino, Italy
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Alessandro Romanel
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Italy
| | - Sabrina Arena
- Department of Oncology, University of Torino, Torino, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Andrea Lunardi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Italy
| |
Collapse
|
7
|
Thomas N, Foukakis T, Willard-Gallo K. The interplay between the immune response and neoadjuvant therapy in breast cancer. Front Oncol 2025; 15:1469982. [PMID: 40421087 PMCID: PMC12104209 DOI: 10.3389/fonc.2025.1469982] [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: 09/12/2024] [Accepted: 04/16/2025] [Indexed: 05/28/2025] Open
Abstract
Treatment of early breast cancer is currently experiencing a rapid evolution because of important insight into tumor subtypes and continuous development and improvement of novel therapeutics. Historically considered non-immunogenic, breast cancer has seen a paradigm shift with increased understanding of immune microenvironment, which have revealed extensive heterogeneity in tumor-associated inflammation. Notably, the more aggressive breast cancer subtypes, including triple-negative and HER2-positive, have exhibited favorable responses to combined chemo-immunotherapy protocols. Neoadjuvant therapy has emerged as the standard of care for these tumors, with pathological complete response used as a surrogate endpoint for long-term clinical outcomes and coincidently expediting new drug approval. The neoadjuvant setting affords a unique opportunity for in vivo treatment response evaluation and effects on the tumor microenvironment. In this review, the predictive and prognostic value of the tumor immune microenvironment before, during, and after treatment across various therapeutic regimens, tailored to distinct breast cancer subtypes, is carefully examined.
Collapse
Affiliation(s)
- Noémie Thomas
- Molecular Immunology Unit, Institut Jules Bordet, Brussel, Belgium
| | - Theodoros Foukakis
- Translational Breast Cancer Research, Department of Oncology-Pathology, Karolinska Institute, Stokholm, Sweden
| | | |
Collapse
|
8
|
Yu Y, Ren W, Mao L, Ouyang W, Hu Q, Yao Q, Tan Y, He Z, Ban X, Hu H, Lin R, Wang Z, Chen Y, Wu Z, Chen K, Ouyang J, Li T, Zhang Z, Liu G, Chen X, Li Z, Duan X, Wang J, Yao H. MRI-based multimodal AI model enables prediction of recurrence risk and adjuvant therapy in breast cancer. Pharmacol Res 2025; 216:107765. [PMID: 40345352 DOI: 10.1016/j.phrs.2025.107765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/06/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Timely intervention and improved prognosis for breast cancer patients rely on early metastasis risk detection and accurate treatment predictions. This study introduces an advanced multimodal MRI and AI-driven 3D deep learning model, termed the 3D-MMR-model, designed to predict recurrence risk in non-metastatic breast cancer patients. We conducted a multicenter study involving 1199 non-metastatic breast cancer patients from four institutions in China, with comprehensive MRI and clinical data retrospectively collected. Our model employed multimodal-data fusion, utilizing contrast-enhanced T1-weighted imaging (T1 + C) and T2-weighted imaging (T2WI) volumes, processed through a modified 3D-UNet for tumor segmentation and a DenseNet121-based architecture for disease-free survival (DFS) prediction. Additionally, we performed RNA-seq analysis to delve further into the relationship between concentrated hotspots within the tumor region and the tumor microenvironment. The 3D-MR-model demonstrated superior predictive performance, with time-dependent ROC analysis yielding AUC values of 0.90, 0.89, and 0.88 for 2-, 3-, and 4-year DFS predictions, respectively, in the training cohort. External validation cohorts corroborated these findings, highlighting the model's robustness across diverse clinical settings. Integration of clinicopathological features further enhanced the model's accuracy, with a multimodal approach significantly improving risk stratification and decision-making in clinical practice. Visualization techniques provided insights into the decision-making process, correlating predictions with tumor microenvironment characteristics. In summary, the 3D-MMR-model represents a significant advancement in breast cancer prognosis, combining cutting-edge AI technology with multimodal imaging to deliver precise and clinically relevant predictions of recurrence risk. This innovative approach holds promise for enhancing patient outcomes and guiding individualized treatment plans in breast cancer care.
Collapse
Affiliation(s)
- Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Cancer Pathogenesis and Precision Diagnosis and Treatment, Joint Big Data Laboratory, Department of Medical Oncology, Shenshan Medical Center, Memorial Hospital of Sun Yat-sen University, Shanwei, China; Institute for AI in Medicine and faculty of Medicine, Macau University of Science and Technology, Taipa, Macao, China; Department of Breast Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Wei Ren
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Luhui Mao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenhao Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiugen Hu
- Department of Radiology, Shunde Hospital, Southern Medical University, Foshan, China
| | - Qinyue Yao
- Cells Vision (Guangzhou) Medical Technology Inc., Guangzhou, China
| | - Yujie Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zifan He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaohua Ban
- Imaging Diagnostic and Interventional Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Huijun Hu
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruichong Lin
- Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa, Macao, China
| | - Zehua Wang
- Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa, Macao, China; UMedEVO and UMedREVO Artificial Intelligence Technology (Guangzhou) Co., Ltd
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Zhuo Wu
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kai Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jie Ouyang
- Department of Breast Surgery, Tungwah Hospital, Dongguan, China
| | - Tang Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zebang Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guoying Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiuxing Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhuo Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaohui Duan
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Jin Wang
- Cells Vision (Guangzhou) Medical Technology Inc., Guangzhou, China.
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
9
|
Mao N, Dai Y, Zhou H, Lin F, Zheng T, Li Z, Yang P, Zhao F, Li Q, Wang W, Liang Y, Xie H, Ma H, Zhang L, Guo Y, Song X, Zhang H, Lu J. A multimodal and fully automated system for prediction of pathological complete response to neoadjuvant chemotherapy in breast cancer. SCIENCE ADVANCES 2025; 11:eadr1576. [PMID: 40305609 PMCID: PMC12042891 DOI: 10.1126/sciadv.adr1576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025]
Abstract
Accurately predicting pathological complete response (pCR) before neoadjuvant chemotherapy (NAC) is crucial for patients with breast cancer. In this study, we developed a multimodal integrated fully automated pipeline system (MIFAPS) in forecasting pCR to NAC, using a multicenter and prospective dataset of 1004 patients with locally advanced breast cancer, incorporating pretreatment magnetic resonance imaging, whole slide image, and clinical risk factors. The results demonstrated that MIFAPS offered a favorable predictive performance in both the pooled external test set [area under the curve (AUC) = 0.882] and the prospective test set (AUC = 0.909). In addition, MIFAPS significantly outperformed single-modality models (P < 0.05). Furthermore, the high deep learning scores were associated with immune-related pathways and the promotion of antitumor cells in the microenvironment during biological basis exploration. Overall, our study demonstrates a promising approach for improving the prediction of pCR to NAC in patients with breast cancer through the integration of multimodal data.
Collapse
Affiliation(s)
- Ning Mao
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing 100053, P. R. China
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, P. R. China
- Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women's Diseases (Yantai Yuhuangding Hospital), Yantai, Shandong 264000, P. R. China
| | - Yi Dai
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P. R. China
| | - Heng Zhou
- School of Information and Electronic Engineering, Shandong Technology and Business University, Yantai, Shandong 264005, P. R. China
| | - Fan Lin
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, P. R. China
| | - Tiantian Zheng
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, P. R. China
- School of Medical Imaging, Binzhou Medical University, Yantai, Shandong 264003, P. R. China
| | - Ziyin Li
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, P. R. China
- School of Medical Imaging, Binzhou Medical University, Yantai, Shandong 264003, P. R. China
| | - Ping Yang
- Department of Pathology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, P. R. China
| | - Feng Zhao
- School of Computer Science and Technology, Shandong Technology and Business University, Yantai, Shandong 264005, P. R. China
| | - Qin Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai 200433, P. R. China
| | - Weiwei Wang
- Department of Medical Imaging, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P. R. China
| | - Yun Liang
- Department of Medical Imaging, Guilin Municipal Hospital of Traditional Chinese Medicine, Guilin, Guangxi 541002, P. R. China
| | - Haizhu Xie
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, P. R. China
| | - Heng Ma
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, P. R. China
| | - Lina Zhang
- Department of Radiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P. R. China
| | - Yuan Guo
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou, Guangdong 510180, P. R. China
| | - Xicheng Song
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women's Diseases (Yantai Yuhuangding Hospital), Yantai, Shandong 264000, P. R. China
| | - Haicheng Zhang
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women's Diseases (Yantai Yuhuangding Hospital), Yantai, Shandong 264000, P. R. China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing 100053, P. R. China
| |
Collapse
|
10
|
Liu W, Chen X, Yang C, Lin Z, Huang X, Zhang Z, Liu J. Preventive effects of xanthohumol in APP/PS1 mice based on multi-omics atlas. Brain Res Bull 2025; 224:111316. [PMID: 40132750 DOI: 10.1016/j.brainresbull.2025.111316] [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: 01/03/2025] [Revised: 03/03/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
Alzheimer's disease (AD) is a complex disease with unknown etiology and pathogenesis. We described a combined analysis of murine proteomics and microbiomics to find potential therapeutic targets of different doses of xanthohumol (Xn), with the goal of providing a biological basis for the treatment of early AD. Xn improved the spatial learning and memory ability of APP/PS1 mice; this was associated with an increased number of newborn neurons in the subgranular zone (SGZ) and dentate gyrus (DG) and a decreased inflammatory response. 108 proteins were significantly changed after 0.5 mg/kg Xn treatment while only 72 proteins changed by 5 mg/kg Xn. Eight significant microbiota were modulated by different doses of Xn at line discriminant analysis (LDA) score 3.0, but only three of which were regulated by 0.5 mg/kg Xn at LDA score 4.0. In addition, Xn treatment could significantly regulate the pathways of neurodegeneration- multiple diseases in the hippocampus and the penicillin and cephalosporin biosynthesis and atrazine degradation pathways in the gut. Interestingly, Nefl protein validated by correlation analysis was found in the common signaling pathway. 0.5 mg/kg Xn was able to reverse the correlation between hippocampal proteins and gut microbiota. Xn treatment significantly improved cognitive function in AD transgenic mice. Different doses of Xn caused significant differences in protein expression and flora composition and abundance, suggesting that the doses of Xn should be selected with caution, and low dose may be better in the prevention of AD.
Collapse
Affiliation(s)
- Wei Liu
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China.
| | - Xiao Chen
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Chen Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Zequn Lin
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Xinfeng Huang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Zhen Zhang
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Jianjun Liu
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| |
Collapse
|
11
|
Zhang M, Zhang Z, Wang J, Yue H, Lou X, Feng Q, Zhang L. Unveiling the Role of PNMA2 in Endometriosis: From Proliferation and Apoptosis to Immunomodulation. J Cell Mol Med 2025; 29:e70576. [PMID: 40323214 PMCID: PMC12051379 DOI: 10.1111/jcmm.70576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 04/02/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
Endometriosis is a chronic disease that jeopardises the quality of life of about 10% of women. The aim of this study was to investigate the expression, function, regulatory mechanism, and relationship with immune cell infiltration of PNMA2 in endometriosis. This study investigates the potential involvement and regulatory mechanisms of PNMA2 in the development of endometriosis through the integration of public data, machine learning, clinical sample transcriptome sequencing, and in vitro cell experiments. Cytological in vitro experiments were conducted to validate the impact of PNMA2 on the modulation of proliferation, migration, apoptosis, and autophagy in 12z cells. Rescue experiments were performed based on the autophagy activator (RAPA) to clarify the regulatory details of PNMA2, apoptosis, and autophagy. The Ciberort algorithm was employed to discern the association between PNMA2 gene expression and more than 20 distinct immune cell infiltration types in endometriosis. The expression of PNMA2 exhibited a notable increase in individuals with endometriosis. Knockdown of PNMA2 inhibited the proliferation and migration of 12z cells. Knockdown of PNMA2 could directly promote apoptosis and could also inhibit autophagy and indirectly promote apoptosis. PNMA2 displayed associations with immune cell infiltration and immunomodulation. The present study demonstrated that the up-regulation of PNMA2 was associated with malignant growth, anti-apoptosis, and immunoregulation of human endometriotic cells. Therefore, PNMA2 may serve as a new diagnostic biomarker and a promising therapeutic target.
Collapse
Affiliation(s)
- Mengjun Zhang
- Department of GynecologyThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zidi Zhang
- Department of GynecologyThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jialin Wang
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Haodi Yue
- Department of Center for Clinical Single Cell BiomedicineHenan Provincial People's Hospital, People's Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xueling Lou
- Department of GynecologyThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Quanling Feng
- Department of GynecologyThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Lindong Zhang
- Department of GynecologyThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
12
|
Dadiani M, Friedlander G, Perry G, Balint-Lahat N, Gilad S, Morzaev-Sulzbach D, Shenoy A, Bossel Ben-Moshe N, Pavlovsky A, Bernstein-Molho R, Domany E, Barshack I, Geiger T, Kaufman B, Gal-Yam EN. Chemoresistome mapping in individual breast cancer patients unravels diversity in dynamic transcriptional adaptation. Mol Oncol 2025. [PMID: 40294066 DOI: 10.1002/1878-0261.70030] [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: 06/30/2024] [Revised: 12/18/2024] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
Nongenetic adaptive resistance to chemotherapy, driven by transcriptional rewiring, is emerging as a significant mechanism in tumor survival. In this study we combined longitudinal transcriptomics with temporal pattern analysis to investigate patient-specific mechanisms underlying acquired resistance in breast cancer. Matched tumor biopsies (pretreatment, posttreatment, and adjacent normal) were collected from breast cancer patients who received neoadjuvant chemotherapy. Transcriptomes were analyzed by longitudinal gene-pattern classification to track patient-specific gene expression alterations that occur during treatment. Our findings reveal that resistance-associated genes were already dysregulated in primary tumors, suggesting the presence of a preexisting drug-tolerant state. While each patient displayed unique resistance-associated gene rewiring, these alterations converged into a limited number of dysregulated functional modules. Notably, patients receiving the same treatment exhibited distinct rewiring of genes and pathways, revealing parallel, individualized routes to resistance. In conclusion, we propose that tumor cells survive chemotherapy by sustaining or amplifying a preexisting drug-tolerant state that circumvents drug action. We suggest that individualized "chemoresistome maps" could identify cancer vulnerabilities and inform personalized therapeutic strategies to overcome or prevent resistance.
Collapse
Affiliation(s)
- Maya Dadiani
- Cancer Research Center, Sheba Medical Center, Ramat Gan, Israel
- The Nehemia Rubin Excellence in Biomedical Research, The TELEM Program, Ramat Gen, Israel
| | - Gilgi Friedlander
- Mantoux Bioinformatics Institute, The Nancy & Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Gili Perry
- Cancer Research Center, Sheba Medical Center, Ramat Gan, Israel
| | | | - Shlomit Gilad
- The Nancy & Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | | | - Anjana Shenoy
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Noa Bossel Ben-Moshe
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot, Israel
| | - Anya Pavlovsky
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel
| | - Rinat Bernstein-Molho
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
- The Suzanne Levy-Gertner Oncogenetics Unit, Sheba Medical Center, Ramat Gan, Israel
- Oncology Institute, Sheba Medical Center, Ramat Gan, Israel
| | - Eytan Domany
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot, Israel
| | - Iris Barshack
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Tamar Geiger
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Bella Kaufman
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
- Oncology Institute, Sheba Medical Center, Ramat Gan, Israel
| | - Einav Nili Gal-Yam
- Oncology Institute, Sheba Medical Center, Ramat Gan, Israel
- The Dr. Pinchas Borenstein Talpiot Medical Leadership Program, Sheba Medical Center, Ramat Gan, Israel
| |
Collapse
|
13
|
Marra A, Morganti S, Pareja F, Campanella G, Bibeau F, Fuchs T, Loda M, Parwani A, Scarpa A, Reis-Filho JS, Curigliano G, Marchiò C, Kather JN. Artificial intelligence entering the pathology arena in oncology: current applications and future perspectives. Ann Oncol 2025:S0923-7534(25)00112-7. [PMID: 40307127 DOI: 10.1016/j.annonc.2025.03.006] [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: 12/18/2024] [Revised: 02/19/2025] [Accepted: 03/07/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Artificial intelligence (AI) is rapidly transforming the fields of pathology and oncology, offering novel opportunities for advancing diagnosis, prognosis, and treatment of cancer. METHODS Through a systematic review-based approach, the representatives from the European Society for Medical Oncology (ESMO) Precision Oncology Working Group (POWG) and international experts identified studies in pathology and oncology that applied AI-based algorithms for tumour diagnosis, molecular biomarker detection, and cancer prognosis assessment. These findings were synthesised to provide a comprehensive overview of current AI applications and future directions in cancer pathology. RESULTS The integration of AI tools in digital pathology is markedly improving the accuracy and efficiency of image analysis, allowing for automated tumour detection and classification, identification of prognostic molecular biomarkers, and prediction of treatment response and patient outcomes. Several barriers for the adoption of AI in clinical workflows, such as data availability, explainability, and regulatory considerations, still persist. There are currently no prognostic or predictive AI-based biomarkers supported by level IA or IB evidence. The ongoing advancements in AI algorithms, particularly foundation models, generalist models and transformer-based deep learning, offer immense promise for the future of cancer research and care. AI is also facilitating the integration of multi-omics data, leading to more precise patient stratification and personalised treatment strategies. CONCLUSIONS The application of AI in pathology is poised to not only enhance the accuracy and efficiency of cancer diagnosis and prognosis but also facilitate the development of personalised treatment strategies. Although barriers to implementation remain, ongoing research and development in this field coupled with addressing ethical and regulatory considerations will likely lead to a future where AI plays an integral role in cancer management and precision medicine. The continued evolution and adoption of AI in pathology and oncology are anticipated to reshape the landscape of cancer care, heralding a new era of precision medicine and improved patient outcomes.
Collapse
Affiliation(s)
- A Marra
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - S Morganti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA; Department of Medicine, Harvard Medical School, Boston, USA; Gerstner Center for Cancer Diagnostics, Broad Institute of MIT and Harvard, Boston, USA
| | - F Pareja
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - G Campanella
- Hasso Plattner Institute for Digital Health, Mount Sinai Medical School, New York, USA; Department of AI and Human Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - F Bibeau
- Department of Pathology, University Hospital of Besançon, Besancon, France
| | - T Fuchs
- Hasso Plattner Institute for Digital Health, Mount Sinai Medical School, New York, USA; Department of AI and Human Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - M Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA; Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, USA
| | - A Parwani
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, USA
| | - A Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy; ARC-Net Research Center, University of Verona, Verona, Italy
| | - J S Reis-Filho
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - G Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - C Marchiò
- Candiolo Cancer Institute, FPO IRCCS, Candiolo, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| | - J N Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany; Department of Medicine I, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
14
|
Wray R, Paverd H, Machado I, Barbieri J, Easita F, Edwards AR, Gallagher FA, Mendichovszky IA, Mitchell TJ, de la Roche M, Shields JD, Ursprung S, Wallis L, Warren AY, Welsh SJ, Crispin-Ortuzar M, Stewart GD, Jones JO. Angiogenic and immune predictors of neoadjuvant axitinib response in renal cell carcinoma with venous tumour thrombus. Nat Commun 2025; 16:3870. [PMID: 40295487 PMCID: PMC12037771 DOI: 10.1038/s41467-025-58436-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/18/2025] [Indexed: 04/30/2025] Open
Abstract
Venous tumour thrombus (VTT), where the primary tumour invades the renal vein and inferior vena cava, affects 10-15% of renal cell carcinoma (RCC) patients. Curative surgery for VTT is high-risk, but neoadjuvant therapy may improve outcomes. The NAXIVA trial demonstrated a 35% VTT response rate after 8 weeks of neoadjuvant axitinib, a VEGFR-directed therapy. However, understanding non-response is critical for better treatment. Here we show that response to axitinib in this setting is characterised by a distinct and predictable set of features. We conduct a multiparametric investigation of samples collected during NAXIVA using digital pathology, flow cytometry, plasma cytokine profiling and RNA sequencing. Responders have higher baseline microvessel density and increased induction of VEGF-A and PlGF during treatment. A multi-modal machine learning model integrating features predict response with an AUC of 0.868, improving to 0.945 when using features from week 3. Key predictive features include plasma CCL17 and IL-12. These findings may guide future treatment strategies for VTT, improving the clinical management of this challenging scenario.
Collapse
Affiliation(s)
- Rebecca Wray
- Early Cancer Institute, University of Cambridge, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Hania Paverd
- Early Cancer Institute, University of Cambridge, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ines Machado
- Early Cancer Institute, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Johanna Barbieri
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Farhana Easita
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Abigail R Edwards
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Ferdia A Gallagher
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Iosif A Mendichovszky
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Thomas J Mitchell
- Early Cancer Institute, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Maike de la Roche
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Jacqueline D Shields
- Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Stephan Ursprung
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Lauren Wallis
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Anne Y Warren
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Sarah J Welsh
- Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | - Mireia Crispin-Ortuzar
- Early Cancer Institute, University of Cambridge, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Grant D Stewart
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - James O Jones
- Department of Oncology, University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
15
|
Zhu R, Eason K, Chin SF, Edwards PAW, Manzano Garcia R, Moulange R, Pan JW, Teo SH, Mukherjee S, Callari M, Caldas C, Sammut SJ, Rueda OM. Detecting homologous recombination deficiency for breast cancer through integrative analysis of genomic data. Mol Oncol 2025. [PMID: 40260608 DOI: 10.1002/1878-0261.70041] [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: 10/05/2024] [Revised: 02/25/2025] [Accepted: 03/30/2025] [Indexed: 04/23/2025] Open
Abstract
Homologous recombination deficiency (HRD) leads to genomic instability, and patients with HRD can benefit from HRD-targeting therapies. Previous studies have primarily focused on identifying HRD biomarkers using data from a single technology. Here we integrated features from different genomic data types, including total copy number (CN), allele-specific copy number (ASCN) and single nucleotide variants (SNV). Using a semi-supervised method, we developed HRD classifiers from 1404 breast tumours across two datasets based on their BRCA1/2 status, demonstrating improved HRD identification when aggregating different data types. Notably, HRD-positive tumours in ER-negative disease showed improved survival post-adjuvant chemotherapy, while HRD status strongly correlated with neoadjuvant treatment response. Furthermore, our analysis of cell lines highlighted a sensitivity to PARP inhibitors, particularly rucaparib, among predicted HRD-positive lines. Exploring somatic mutations outside BRCA1/2, we confirmed variants in several genes associated with HRD. Our method for HRD classification can adapt to different data types or resolutions and can be used in various scenarios to help refine patient selection for HRD-targeting therapies that might lead to better clinical outcomes.
Collapse
Affiliation(s)
- Rong Zhu
- School of Mathematics and Statistics, Beijing Institute of Technology, Beijing, China
- MRC Biostatistics Unit, University of Cambridge, UK
| | - Katherine Eason
- Cancer Research UK Cambridge Institute, University of Cambridge, UK
| | - Suet-Feung Chin
- Cancer Research UK Cambridge Institute, University of Cambridge, UK
| | | | | | | | | | | | - Sach Mukherjee
- MRC Biostatistics Unit, University of Cambridge, UK
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- University of Bonn, Bonn, Germany
| | | | - Carlos Caldas
- School of Clinical Medicine, University of Cambridge, UK
| | - Stephen-John Sammut
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | | |
Collapse
|
16
|
Guan J, Fan M, Li L. MVNMF: Multiview nonnegative matrix factorization for radio-multigenomic analysis in breast cancer prognosis. Med Image Anal 2025; 103:103566. [PMID: 40288334 DOI: 10.1016/j.media.2025.103566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 02/16/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
Radiogenomic research provides a deeper understanding of breast cancer biology by investigating the correlations between imaging phenotypes and genetic data. However, current radiogenomic research primarily focuses on the correlation between imaging phenotypes and single-genomic data (e.g., gene expression data), overlooking the potential of multi-genomics data to unveil more nuances in cancer characterization. To this end, we propose a multiview nonnegative matrix factorization (MVNMF) method for the radio-multigenomic analysis that identifies dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) features associated with multi-genomics data, including DNA copy number alterations, mutations, and mRNAs, each of which is independently predictive of cancer outcomes. MVNMF incorporates subspace learning and multiview regularization into a unified model to simultaneously select features and explore correlations. Subspace learning is utilized to identify representative radiomic features crucial for tumor analysis, while multiview regularization enables the learning of the correlation between the identified radiomic features and multi-genomics data. Experimental results showed that, for overall survival prediction in breast cancer, MVNMF classified patients into two distinct groups characterized by significant differences in survival (p = 0.0012). Furthermore, it achieved better performance with a C-index of 0.698 compared to the method without considering any genomics data (C-index = 0.528). MVNMF is an effective framework for identifying radiomic features linked to multi-genomics data, which improves its predictive power and provides a better understanding of the biological mechanisms underlying observed phenotypes. MVNMF offers a novel framework for prognostic prediction in breast cancer, with the potential to catalyze further radiogenomic/radio-multigenomic studies.
Collapse
Affiliation(s)
- Jian Guan
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou 310018, China; College of Mathematics and Data Science, Minjiang University, Fuzhou 350121, China
| | - Ming Fan
- Institute of Intelligent Biomedicine, Hangzhou Dianzi University, Hangzhou 310018, China.
| | - Lihua Li
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou 310018, China; Institute of Intelligent Biomedicine, Hangzhou Dianzi University, Hangzhou 310018, China.
| |
Collapse
|
17
|
Mousavi R, Mustafa Ali MK, Lobo D. Discovery of Dynamic Models for AML Disease Progression from Longitudinal Multi-Modal Clinical Data Using Explainable Machine Learning. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.07.25325267. [PMID: 40297459 PMCID: PMC12036371 DOI: 10.1101/2025.04.07.25325267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Acute Myeloid Leukemia (AML) is a complex and heterogeneous disease identified by severe clinical progression, fast cellular proliferation, and often high mortality rates. Incorporating diverse longitudinal information on patients' medical histories is essential for developing effective disease predictive models applicable to both research and clinical settings. Here, we present a robust methodology for discovering dynamic predictive models to elucidate AML disease progression dynamics from a novel longitudinal multimodal clinical dataset of patients diagnosed with AML. The clinical dataset was analyzed to reveal the main clinical, genetic, and treatment features modulating disease progression. To discover mathematical models-including interactions, parameters, and nodes-predictive of AML progression, we present an explainable machine learning algorithm based on high-performance evolutionary computation. The results demonstrate that the predictive methodology could accurately estimate the clinical dynamics of AML progression in terms of blast percentages for both training and novel patients. This study demonstrates that the developed explainable machine learning approach can successfully predict AML progression by leveraging the heterogeneous and longitudinal dynamics of patients' clinical data. More importantly, this methodology shows significant potential for application in modeling the progression dynamics of other acute diseases, providing a flexible and adaptable framework for advancing clinical and translational research.
Collapse
Affiliation(s)
- Reza Mousavi
- Department of Biological Sciences, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Moaath K. Mustafa Ali
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Daniel Lobo
- Department of Biological Sciences, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center and Center for Stem Cell Biology & Regenerative Medicine, University of Maryland, School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, USA
| |
Collapse
|
18
|
Jin Y, Zhao M, Su T, Fan Y, Ouyang Z, Lv F. Comparing Random Survival Forests and Cox Regression for Nonresponders to Neoadjuvant Chemotherapy Among Patients With Breast Cancer: Multicenter Retrospective Cohort Study. J Med Internet Res 2025; 27:e69864. [PMID: 40198909 PMCID: PMC12015342 DOI: 10.2196/69864] [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: 12/10/2024] [Revised: 02/19/2025] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Breast cancer is one of the most common malignancies among women worldwide. Patients who do not achieve a pathological complete response (pCR) or a clinical complete response (cCR) post-neoadjuvant chemotherapy (NAC) typically have a worse prognosis compared to those who do achieve these responses. OBJECTIVE This study aimed to develop and validate a random survival forest (RSF) model to predict survival risk in patients with breast cancer who do not achieve a pCR or cCR post-NAC. METHODS We analyzed patients with no pCR/cCR post-NAC treated at the First Affiliated Hospital of Chongqing Medical University from January 2019 to 2023, with external validation in Duke University and Surveillance, Epidemiology, and End Results (SEER) cohorts. RSF and Cox regression models were compared using the time-dependent area under the curve (AUC), the concordance index (C-index), and risk stratification. RESULTS The study cohort included 306 patients with breast cancer, with most aged 40-60 years (204/306, 66.7%). The majority had invasive ductal carcinoma (290/306, 94.8%), with estrogen receptor (ER)+ (182/306, 59.5%), progesterone receptor (PR)- (179/306, 58.5%), and human epidermal growth factor receptor 2 (HER2)+ (94/306, 30.7%) profiles. Most patients presented with T2 (185/306, 60.5%), N1 (142/306, 46.4%), and M0 (295/306, 96.4%) staging (TNM meaning "tumor, node, metastasis"), with 17.6% (54/306) experiencing disease progression during a median follow-up of 25.9 months (IQR 17.2-36.3). External validation using Duke (N=94) and SEER (N=2760) cohorts confirmed consistent patterns in age (40-60 years: 59/94, 63%, vs 1480/2760, 53.6%), HER2+ rates (26/94, 28%, vs 935/2760, 33.9%), and invasive ductal carcinoma prevalence (89/94, 95%, vs 2506/2760, 90.8%). In the internal cohort, the RSF achieved significantly higher time-dependent AUCs compared to Cox regression at 1-year (0.811 vs 0.763), 3-year (0.834 vs 0.783), and 5-year (0.810 vs 0.771) intervals (overall C-index: 0.803, 95% CI 0.747-0.859, vs 0.736, 95% CI 0.673-0.799). External validation confirmed robust generalizability: the Duke cohort showed 1-, 3-, and 5-year AUCs of 0.912, 0.803, and 0.776, respectively, while the SEER cohort maintained consistent performance with AUCs of 0.771, 0.729, and 0.702, respectively. Risk stratification using the RSF identified 25.8% (79/306) high-risk patients and a significantly reduced survival time (P<.001). Notably, the RSF maintained improved net benefits across decision thresholds in decision curve analysis (DCA); similar results were observed in external studies. The RSF model also showed promising performance across different molecular subtypes in all datasets. Based on the RSF predicted scores, patients were stratified into high- and low-risk groups, with notably poorer survival outcomes observed in the high-risk group compared to the low-risk group. CONCLUSIONS The RSF model, based solely on clinicopathological variables, provides a promising tool for identifying high-risk patients with breast cancer post-NAC. This approach may facilitate personalized treatment strategies and improve patient management in clinical practice.
Collapse
Affiliation(s)
- Yudi Jin
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min Zhao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tong Su
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yanjia Fan
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zubin Ouyang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fajin Lv
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
19
|
Xu Y, Wang T, Liang X, Yang J, Zhang Y, Bao S. Global research trends and focus on immunotherapy for endometrial cancer: a comprehensive bibliometric insight and visualization analysis (2012-2024). Front Immunol 2025; 16:1571800. [PMID: 40264788 PMCID: PMC12011754 DOI: 10.3389/fimmu.2025.1571800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Background This study conducted a novel systematic bibliometric and visualization analysis of global literature on immunotherapy for endometrial cancer (EC) to explore dynamic trends, research hotspots, and emerging topics, providing valuable references for future research. Methods Articles and reviews on EC immunotherapy published between 2012 and August 2024 were retrieved from the Web of Science Core Collection (WoSCC). Bibliometric tools, CiteSpace and VOSviewer, were used to analyze clustering patterns and research dynamics. Results A total of 861 articles were contributed by 5,331 authors from 1,392 institutions across 58 countries or regions, involving 1,823 keywords. China demonstrated outstanding performance in this field, contributing over 40% of the total publications and ranking first in publication volume. However, the total citation counts for publications from China lags that of the United States, highlighting the latter's leading position and areas for further improvement in China's research efforts. The University of Texas Medical Anderson Cancer Center and Nanjing Medical University were the two institutions with the highest number of publications. In terms of authorship, research teams led by Bosse, Tjalling, and Creutzberg, Carien L made significant contributions to advancing the field. Among individual publications, the work by Talhouk et al. achieved the highest average annual citation count of 70.88, demonstrating its profound impact. In terms of journals, Gynecologic Oncology emerged as a pivotal academic platform, publishing numerous articles and achieving the highest co-citation frequency. Additionally, Frontiers in Oncology, Frontiers in Immunology, and Frontiers in Genetics have become some of the most active and rapidly developing journals in recent years. Research hotspots are concentrated on themes such as the "Tumor Immune Microenvironment", "Immune Checkpoint Inhibitors", and "Targeted Therapy". Recent trends and frontier research focus on the combined application of immune checkpoint inhibitors with other therapies, research on the application of nanotechnology in immunotherapy, and the integration of artificial intelligence to enhance precision medicine. Additionally, efforts are increasingly directed toward advancing various immunotherapy strategies from basic research to clinical applications. Conclusions This comprehensive analysis reveals rapid advancements and significant potential in EC immunotherapy. Strengthening international collaboration and addressing barriers in the translation of research to clinical practice will drive further progress in this promising field.
Collapse
Affiliation(s)
- Yachen Xu
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Tao Wang
- School of Public Health, Hainan Medical University, Haikou, China
| | - Xiaojing Liang
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Jie Yang
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Yuxiang Zhang
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Shan Bao
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| |
Collapse
|
20
|
Wu C, Lima EABF, Stowers CE, Xu Z, Yam C, Son JB, Ma J, Rauch GM, Yankeelov TE. MRI-based digital twins to improve treatment response of breast cancer by optimizing neoadjuvant chemotherapy regimens. NPJ Digit Med 2025; 8:195. [PMID: 40195521 PMCID: PMC11976917 DOI: 10.1038/s41746-025-01579-1] [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: 08/13/2024] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
We developed a practical framework to construct digital twins for predicting and optimizing triple-negative breast cancer (TNBC) response to neoadjuvant chemotherapy (NAC). This study employed 105 TNBC patients from the ARTEMIS trial (NCT02276443, registered on 10/21/2014) who received Adriamycin/Cytoxan (A/C)-Taxol (T). Digital twins were established by calibrating a biology-based mathematical model to patient-specific MRI data, which accurately predicted pathological complete response (pCR) with an AUC of 0.82. We then used each patient's twin to theoretically optimize outcome by identifying their optimal A/C-T schedule from 128 options. The patient-specifically optimized treatment yielded a significant improvement in pCR rate of 20.95-24.76%. Retrospective validation was conducted by virtually treating the twins with AC-T schedules from historical trials and obtaining identical observations on outcomes: bi-weekly A/C-T outperforms tri-weekly A/C-T, and weekly/bi-weekly T outperforms tri-weekly T. This proof-of-principle study demonstrates that our digital twin framework provides a practical methodology to identify patient-specific TNBC treatment schedules.
Collapse
Affiliation(s)
- Chengyue Wu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, USA.
| | - Ernesto A B F Lima
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, USA
- Texas Advanced Computing Center, The University of Texas at Austin, Austin, TX, USA
| | - Casey E Stowers
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, USA
| | - Zhan Xu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Clinton Yam
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jong Bum Son
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jingfei Ma
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gaiane M Rauch
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas E Yankeelov
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX, USA
- Department of Oncology, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
21
|
Diop M, Davidson BR, Fragiadakis GK, Sirota M, Gaudillière B, Combes AJ. Single-cell omics technologies - Fundamentals on how to create single-cell looking glasses for reproductive health. Am J Obstet Gynecol 2025; 232:S1-S20. [PMID: 40253074 PMCID: PMC12090843 DOI: 10.1016/j.ajog.2024.08.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 07/18/2024] [Accepted: 08/24/2024] [Indexed: 04/21/2025]
Abstract
Over the last decade, in line with the goals of precision medicine to offer individualized patient care, various single-cell technologies measuring gene and proteomic expression in various tissues have rapidly advanced to study health and disease at the single cell level. Precisely understanding cell composition, position within tissues, signaling pathways, and communication can reveal insights into disease mechanisms and systemic changes during development, pregnancy, and gynecologic disorders across the lifespan. Single-cell technologies dissect the complex cellular compositions of reproductive tract tissues, providing insights into mechanisms behind reproductive tract dysfunction which impact wellness and quality of life. These technologies aim to understand basic tissue and organ functions and, clinically, to develop novel diagnostics, early disease biomarkers, and cell-targeted therapies for currently suboptimally-treated disorders. Increasingly, they are applied to pregnancy and pregnancy disorders, gynecologic malignancies, and uterine and ovarian physiology and aging, which are discussed in more detail in manuscripts in this special issue of AJOG. Here, we review recent applications of single-cell technologies to the study of gynecologic disorders and systemic biological adaptations during fetal development, pregnancy, and across a woman's lifespan. We discuss sequencing- and proteomic-based single-cell methods, as well as spatial transcriptomics and high-dimensional proteomic imaging, describing each technology's mechanism, workflow, quality control, and highlighting specific benefits, drawbacks, and utility in the context of reproductive medicine. We consider analytical methods for the high-dimensional single-cell data generated, highlighting statistical constraints and recent computational techniques for downstream clinical translation. Overall, current and evolving single-cell "looking glasses", or perspectives, have the potential to transform fundamental understanding of women's health and reproductive disorders and alter the trajectory of clinical practice and patient outcomes in the future.
Collapse
Affiliation(s)
- Maïgane Diop
- Program in Immunology, Stanford University School of Medicine, Stanford, CA; Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA
| | | | - Gabriela K Fragiadakis
- UCSF CoLabs, University of California, San Francisco, CA; Bakar ImmunoX Initiative, University of California, San Francisco, CA; Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA.
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA; Department of Pediatrics, University of California, San Francisco, CA.
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA.
| | - Alexis J Combes
- UCSF CoLabs, University of California, San Francisco, CA; Department of Pathology, University of California, San Francisco, CA; Bakar ImmunoX Initiative, University of California, San Francisco, CA; Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA.
| |
Collapse
|
22
|
Llinas-Bertran A, Butjosa-Espín M, Barberi V, Seoane JA. Multimodal data integration in early-stage breast cancer. Breast 2025; 80:103892. [PMID: 39922065 PMCID: PMC11973824 DOI: 10.1016/j.breast.2025.103892] [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: 10/10/2024] [Revised: 12/13/2024] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
The use of biomarkers in breast cancer has significantly improved patient outcomes through targeted therapies, such as hormone therapy anti-Her2 therapy and CDK4/6 or PARP inhibitors. However, existing knowledge does not fully encompass the diverse nature of breast cancer, particularly in triple-negative tumors. The integration of multi-omics and multimodal data has the potential to provide new insights into biological processes, to improve breast cancer patient stratification, enhance prognosis and response prediction, and identify new biomarkers. This review presents a comprehensive overview of the state-of-the-art multimodal (including molecular and image) data integration algorithms developed and with applicability to breast cancer stratification, prognosis, or biomarker identification. We examined the primary challenges and opportunities of these multimodal data integration algorithms, including their advantages, limitations, and critical considerations for future research. We aimed to describe models that are not only academically and preclinically relevant, but also applicable to clinical settings.
Collapse
Affiliation(s)
- Arnau Llinas-Bertran
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Maria Butjosa-Espín
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Vittoria Barberi
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jose A Seoane
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
23
|
Zhang HW, Wang YR, Li J, Huang W, Xu B, Pang HW, Jiang CL. Predicting Radiation Esophagitis in Patients Undergoing Synchronous Boost Radiotherapy Post-Breast-Conserving Surgery. Dose Response 2025; 23:15593258251335802. [PMID: 40297669 PMCID: PMC12033885 DOI: 10.1177/15593258251335802] [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: 10/04/2024] [Revised: 02/28/2025] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
This study constructed a predictive model for occurrence of radiation esophagitis during breast-cancer radiotherapy. 308 breast-cancer patients were analyzed. Lasso regression identified crucial variables that were further integrated into a radiation esophagitis risk score, which was used to segregate patients into high- and low-risk groups. A nomogram model was designed for clinical applicability. Training and validations were performed to assess robustness and generalizability of proposed models, employing C-index, AUCs, calibration curves, and decision curves. SHAP algorithm was used for model interpretation, offering insights into the major contributory factors. Seven significant variables were identified by Lasso regression. C-indexes of nomograms of individual clinical variables and risk score were 0.795 and 0.784, respectively, exhibiting strong predictive ability. In internal validation, AUCs for risk score, nomogram, and logistic models were 0.784, 0.795, and 0.812, respectively. Calibration curves showed a close fit between predicted and observed outcomes across models. Decision curve analysis indicated logistic model's superior clinical utility when the risk threshold was above 0.2. SHAP interpretation emphasized radiation dose, pruritus, molecular type, and hepatic dysfunction as top contributory factors for radiation esophagitis. Models based on interpretable machine learning offer an intuitive tool to assess risk of radiation esophagitis in breast-cancer radiotherapy.
Collapse
Affiliation(s)
- Huai-wen Zhang
- Department of Radiation Oncology, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yi-ren Wang
- School of Nursing, Southwest Medical University, Luzhou, China
| | - Jingao Li
- Department of Radiation Oncology, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Wei Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Bin Xu
- Department of Radiation Oncology, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Hao-wen Pang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chun-ling Jiang
- Department of Radiation Oncology, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Medical College of Nanchang University, China
| |
Collapse
|
24
|
Feng K, Yi Z, Xu B. Artificial Intelligence and Breast Cancer Management: From Data to the Clinic. CANCER INNOVATION 2025; 4:e159. [PMID: 39981497 PMCID: PMC11840326 DOI: 10.1002/cai2.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 02/22/2025]
Abstract
Breast cancer (BC) remains a significant threat to women's health worldwide. The oncology field had an exponential growth in the abundance of medical images, clinical information, and genomic data. With its continuous advancement and refinement, artificial intelligence (AI) has demonstrated exceptional capabilities in processing intricate multidimensional BC-related data. AI has proven advantageous in various facets of BC management, encompassing efficient screening and diagnosis, precise prognosis assessment, and personalized treatment planning. However, the implementation of AI into precision medicine and clinical practice presents ongoing challenges that necessitate enhanced regulation, transparency, fairness, and integration of multiple clinical pathways. In this review, we provide a comprehensive overview of the current research related to AI in BC, highlighting its extensive applications throughout the whole BC cycle management and its potential for innovative impact. Furthermore, this article emphasizes the significance of constructing patient-oriented AI algorithms. Additionally, we explore the opportunities and potential research directions within this burgeoning field.
Collapse
Affiliation(s)
- Kaixiang Feng
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Zongbi Yi
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
25
|
Li Q, Liu H. Investigating the Prognostic Role of Telomerase-Related Cellular Senescence Gene Signatures in Breast Cancer Using Machine Learning. Biomedicines 2025; 13:826. [PMID: 40299459 PMCID: PMC12024799 DOI: 10.3390/biomedicines13040826] [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: 01/26/2025] [Revised: 03/24/2025] [Accepted: 03/29/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Telomeres and cellular senescence are critical biological processes implicated in cancer development and progression, including breast cancer, through their influence on genomic stability and modulation of the tumor microenvironment. Methods: This study integrated bulk RNA sequencing and single-cell RNA sequencing (scRNA-seq) data to establish a gene signature associated with telomere maintenance and cellular senescence for prognostic prediction in breast cancer. Telomere-related genes (TEGs) and senescence-associated genes were curated from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. A comprehensive machine learning framework incorporating 101 algorithmic combinations across 10 survival modeling approaches, including random survival forests and ridge regression, was employed to develop a robust prognostic model. Results: A set of 19 key telomere- and senescence-related genes was identified as the optimal prognostic signature. The model demonstrated strong predictive accuracy and was successfully validated in multiple independent cohorts. Functional enrichment analyses indicated significant associations with immune responses and aging-related pathways. Single-cell transcriptomic analysis revealed marked cellular heterogeneity, identifying distinct subpopulations (fibroblasts and immune cells) with divergent risk scores and biological pathway activity. Additionally, pseudo-time trajectory analysis and intercellular communication mapping provided insights into the dynamic evolution of the tumor microenvironment. Immunohistochemical (IHC) validation using data from the Human Protein Atlas confirmed differential protein expression between normal and tumor tissues for several of the selected genes, reinforcing their biological relevance and clinical utility. Conclusions: This study presents a novel 19-gene telomere- and senescence-associated signature with strong prognostic value in breast cancer. These findings enhance our understanding of tumor heterogeneity and may inform precision oncology approaches and future therapeutic strategies.
Collapse
Affiliation(s)
| | - Hongde Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China;
| |
Collapse
|
26
|
Wu Y, Shi Y, Luo Z, Zhou X, Chen Y, Song X, Liu S. Spatial multi-omics analysis of tumor-stroma boundary cell features for predicting breast cancer progression and therapy response. Front Cell Dev Biol 2025; 13:1570696. [PMID: 40206396 PMCID: PMC11979139 DOI: 10.3389/fcell.2025.1570696] [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: 02/04/2025] [Accepted: 03/05/2025] [Indexed: 04/11/2025] Open
Abstract
Background The tumor boundary of breast cancer represents a highly heterogeneous region. In this area, the interactions between malignant and non-malignant cells influence tumor progression, immune evasion, and drug resistance. However, the spatial transcriptional profile of the tumor boundary and its role in the prognosis and treatment response of breast cancer remain unclear. Method Utilizing the Cottrazm algorithm, we reconstructed the intricate boundaries and identified differentially expressed genes (DEGs) associated with these regions. Cell-cell co-positioning analysis was conducted using SpaCET, which revealed key interactions between tumor-associated macrophage (TAMs) and cancer-associated fibroblasts (CAFs). Additionally, Lasso regression analysis was employed to develop a malignant body signature (MBS), which was subsequently validated using the TCGA dataset for prognosis prediction and treatment response assessment. Results Our research indicates that the tumor boundary is characterized by a rich reconstruction of the extracellular matrix (ECM), immunomodulatory regulation, and the epithelial-to-mesenchymal transition (EMT), underscoring its significance in tumor progression. Spatial colocalization analysis reveals a significant interaction between CAFs and M2-like tumor-associated macrophage (TAM), which contributes to immune exclusion and drug resistance. The MBS score effectively stratifies patients into high-risk groups, with survival outcomes for patients exhibiting high MBS scores being significantly poorer. Furthermore, drug sensitivity analysis demonstrates that high-MB tumors had poor response to chemotherapy strategies, highlighting the role of the tumor boundary in modulating therapeutic efficacy. Conclusion Collectively, we investigate the spatial transcription group and bulk data to elucidate the characteristics of tumor boundary molecules in breast cancer. The CAF-M2 phenotype emerges as a critical determinant of immunosuppression and drug resistance, suggesting that targeting this interaction may improve treatment responses. Furthermore, the MBS serves as a novel prognostic tool and offers potential strategies for guiding personalized treatment approaches in breast cancer.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Youyang Shi
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhanyang Luo
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xiqiu Zhou
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yonghao Chen
- West China Hospital of Sichuan University, Chengdu, China
| | - Xiaoyun Song
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sheng Liu
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
27
|
Zhu Q, Balasubramanian A, Asirvatham JR, Chatterjee M, Piyarathna B, Kaur J, Mohamed N, Wu L, Wang S, Pourfarrokh N, Binsol PD, Bhargava M, Rasaily U, Xu Y, Zheng J, Jebakumar D, Rao A, Gutierrez C, Omilian A, Morrison C, Das GM, Ambrosone C, Seeley EH, Chen SH, Li Y, Chang E, Li X, Baker E, Aneja R, Zhang XHF, Sreekumar A. Integrative spatial omics reveals distinct tumor-promoting multicellular niches and immunosuppressive mechanisms in Black American and White American patients with TNBC. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.17.585428. [PMID: 38562769 PMCID: PMC10983891 DOI: 10.1101/2024.03.17.585428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Racial disparities in the clinical outcomes of triple-negative breast cancer (TNBC) have been well-documented, but the underlying biological mechanisms remain poorly understood. To investigate these disparities, we employed a multi-omic approach integrating imaging mass cytometry and spatial transcriptomics to characterize the tumor microenvironment (TME) in self-identified Black American (BA) and White American (WA) TNBC patients. Our analysis revealed that the TME in BA patients is marked by a network of endothelial cells, macrophages, and mesenchymal-like cells, which correlates with reduced patient survival. In contrast, the WA TNBC microenvironment is enriched in T-cells and neutrophils, indicative of T-cell exhaustion and suppressed immune responses. Ligand-receptor and pathway analyses further demonstrated that BA TNBC tumors exhibit a relatively "immune-cold" profile, while WA TNBC tumors display features of an "inflamed" TME, suggesting the evolution of a unique immunosuppressive mechanism. These findings provide insight into racially distinct tumor-promoting and immunosuppressive microenvironments, which may contribute to the observed differences in clinical outcomes among BA and WA TNBC patients. Statement of Significance This study identifies distinct tumor microenvironment (TME) profiles in Black and White American TNBC patients, providing new insights into the biological mechanisms driving outcome disparities. Our findings highlight the role of the tumor-endothelial-macrophage niche in these disparities, offering a potential therapeutic target for race-inclusive strategies aimed at improving clinical outcomes. By revealing racial differences in treatment response profiles, this work underscores the necessity for tailored therapies in TNBC. These insights lay the groundwork for the development of inclusive, precision-driven treatment approaches that may help mitigate racial disparities and enhance patient outcomes.
Collapse
|
28
|
Macias RIR, Roessler S, Verstegen MMA. Deciphering the spatial tumor microenvironment in intrahepatic cholangiocarcinoma. Hepatology 2025:01515467-990000000-01212. [PMID: 40127117 DOI: 10.1097/hep.0000000000001322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 03/26/2025]
Affiliation(s)
- Rocio I R Macias
- Department of Physiology and Pharmacology, Laboratory of Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, National Institute for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), IBSAL, Salamanca, Spain
| | - Stephanie Roessler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC Transplantation Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
29
|
Rawashdeh B, Al-abdallat H, Arpali E, Thomas B, Dunn TB, Cooper M. Machine learning in solid organ transplantation: Charting the evolving landscape. World J Transplant 2025; 15:99642. [PMID: 40104197 PMCID: PMC11612896 DOI: 10.5500/wjt.v15.i1.99642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/17/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Machine learning (ML), a major branch of artificial intelligence, has not only demonstrated the potential to significantly improve numerous sectors of healthcare but has also made significant contributions to the field of solid organ transplantation. ML provides revolutionary opportunities in areas such as donor-recipient matching, post-transplant monitoring, and patient care by automatically analyzing large amounts of data, identifying patterns, and forecasting outcomes. AIM To conduct a comprehensive bibliometric analysis of publications on the use of ML in transplantation to understand current research trends and their implications. METHODS On July 18, a thorough search strategy was used with the Web of Science database. ML and transplantation-related keywords were utilized. With the aid of the VOS viewer application, the identified articles were subjected to bibliometric variable analysis in order to determine publication counts, citation counts, contributing countries, and institutions, among other factors. RESULTS Of the 529 articles that were first identified, 427 were deemed relevant for bibliometric analysis. A surge in publications was observed over the last four years, especially after 2018, signifying growing interest in this area. With 209 publications, the United States emerged as the top contributor. Notably, the "Journal of Heart and Lung Transplantation" and the "American Journal of Transplantation" emerged as the leading journals, publishing the highest number of relevant articles. Frequent keyword searches revealed that patient survival, mortality, outcomes, allocation, and risk assessment were significant themes of focus. CONCLUSION The growing body of pertinent publications highlights ML's growing presence in the field of solid organ transplantation. This bibliometric analysis highlights the growing importance of ML in transplant research and highlights its exciting potential to change medical practices and enhance patient outcomes. Encouraging collaboration between significant contributors can potentially fast-track advancements in this interdisciplinary domain.
Collapse
Affiliation(s)
- Badi Rawashdeh
- Division of Transplant Surgery, Medical College of Wisconsin, Milwaukee, WI 53202, United States
| | | | - Emre Arpali
- Division of Transplant Surgery, Medical College of Wisconsin, Milwaukee, WI 53202, United States
| | - Beje Thomas
- Department of Nephrology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Ty B Dunn
- Division of Transplant Surgery, Medical College of Wisconsin, Milwaukee, WI 53202, United States
| | - Matthew Cooper
- Division of Transplant Surgery, Medical College of Wisconsin, Milwaukee, WI 53202, United States
| |
Collapse
|
30
|
Mu M, Wang G, Chen B, Li H, Feng C, Fan R, Chen N, Han B, Tong A, Zou B, Guo G. Decomposable STING nanoagonist-amplified oncolytic virotherapy through remodeling the immunosuppressive microenvironment of triple-negative breast cancer. J Mater Chem B 2025; 13:3685-3699. [PMID: 39981850 DOI: 10.1039/d4tb02565b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Oncolytic viruses (OVs) are promising for cancer treatment as they specifically replicate in tumor cells. However, the systemic delivery of OVs still faces the challenges of poor tumor targeting, short circulation periods, and limited lytic efficacy. Herein, an OV-concealed targeting nanoagonist (OV-MnO2/HE) was prepared to enhance the delivery of OVs to triple-negative breast cancer (TNBC) via intravenous administration. Decomposable MnO2 biomineral shells covered the surface antigens of OVs to prevent their clearance after systemic administration. The targeting materials of HA-EGCG (HE) enhanced intratumoral accumulation via active targeting. After entering tumors, OV-MnO2/HE readily released Mn2+ and OVs, which could enhance the number of CD4+/CD8+ T cells and maturation dendritic cells (DCs) due to the synergetic effect of the STING pathway and OVs, thereby activating the immune response, resulting in the significant inhibition of TNBC growth. This work highlights the potential of the STING agonist in enhancing the antitumor efficacy of OVs and provides a potent platform for TNBC therapy.
Collapse
Affiliation(s)
- Min Mu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Guoqing Wang
- Department of Ophthalmology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bo Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Hui Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Chenqian Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Rangrang Fan
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Nianyong Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Bo Han
- Key Laboratory of Xinjiang Endemic Phytomedicine Resources Ministry of Education, Shihezi University College of Pharmacy, Shihezi, 832002, China
| | - Aiping Tong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Bingwen Zou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Gang Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
31
|
Yang H, Chen Y, Zhao A, Rao X, Li L, Li Z. Development of a machine learning-based predictive model for maxillary sinus cysts and exploration of clustering patterns. Head Face Med 2025; 21:17. [PMID: 40069749 PMCID: PMC11900490 DOI: 10.1186/s13005-025-00492-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND AND OBJECTIVE There are still many controversies about the factors influencing maxillary sinus cysts and their clinical management. This study aims to construct a prediction model of maxillary sinus cyst and explore its clustering pattern by cone beam computerized tomography (CBCT) technique and machine learning (ML) method to provide a theoretical basis for the prevention and clinical management of maxillary sinus cyst. METHODS In this study, 6000 CBCT images of maxillary sinus from 3093 patients were evaluated to document the possible influencing factors of maxillary sinus cysts, including gender, age, odontogenic factors, and anatomical factors. First, the characteristic variables were screened by multiple statistical methods, and ML methods were applied to construct a prediction model for maxillary sinus cysts. Second, the model was interpreted based on the SHapley Additive exPlanations (SHAP) values, and the risk of maxillary sinus cysts was predicted by generating a web page calculator. Finally, the K-mean clustering algorithm further identified risk factors for maxillary sinus cysts. RESULTS By comparing the various metrics in the training and test sets of multiple ML models, eXtreme Gradient Boosting (XGBoost) is the best model. The average area under curve (AUC) values of the XGBoost model in the training, validation, and test sets, respectively, are 0.939, 0.923, and 0.921, which indicates its excellent classification and discrimination ability. The cluster analysis model further categorized maxillary sinus cysts into high-risk and low-risk groups, with apical lesions, severe periodontitis, and age ≥ 53 as high-risk factors for maxillary sinus cysts. CONCLUSION These findings provide valuable insights into the etiology and risk stratification of maxillary sinus cysts, offering a theoretical basis for their prevention and clinical management. The integration of CBCT imaging and ML techniques holds the potential for prevention and personalized treatment strategies of maxillary sinus cysts.
Collapse
Affiliation(s)
- Haoran Yang
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Stomatology, Kunming, Yunnan, China
- Kunming Medical University Haiyuan College, Kunming, Yunnan, China
| | - Yuxiang Chen
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Stomatology, Kunming, Yunnan, China
| | - Anna Zhao
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Stomatology, Kunming, Yunnan, China
| | - Xianqi Rao
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Stomatology, Kunming, Yunnan, China
| | - Lin Li
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Stomatology, Kunming, Yunnan, China
| | - Ziliang Li
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China.
- Yunnan Provincial Key Laboratory of Stomatology, Kunming, Yunnan, China.
| |
Collapse
|
32
|
Zhang J, Yang Y, Shang M, Guo L, Zhang D, Du L, for the Alzheimer’s Disease Neuroimaging Initiative. Mutual-assistance learning for trustworthy biomarker discovery and disease prediction. Brief Bioinform 2025; 26:bbaf178. [PMID: 40254831 PMCID: PMC12009715 DOI: 10.1093/bib/bbaf178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/23/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Integrating and analyzing multiple omics datasets, such as genomics, environmental influences, and imaging endophenotypes, has yielded an abundance of candidate biomarkers. However, translating such findings into beneficial clinical knowledge for disease prediction remains challenging. This becomes even more challenging when studying interpretable high-order feature interactions such as gene-environment interaction (G$\times $E) to understand the etiology. To fill this gap, we draw on the idea of mutual-assistance (MA) learning and accordingly propose a fresh and powerful scheme, referred to as mutual-assistance causal biomarker discovery and stable disease prediction approach (MA-CBxDP). Specifically, we design an interpretable bi-directional mapping framework, integrated with a causal feature interaction module, to extract co-expression patterns across different modalities and identify trustworthy biomarkers including G$\times $E. A cooperative prediction module is further incorporated to ensure accurate diagnosis and identification of causal effects for pathogenesis. Importantly, biomarker discovery and disease prediction can mutually reinforce each other, helping to provide novel insights into chronic diseases. Furthermore, in light of the large computational burden incurred by the high-dimensional interactions, we devise a rapid strategy and extend it to a more practical but challenging chromosome-wide setting. We conduct extensive experiments on two databases under three tasks, i.e. multimodal correlation, disease diagnosis, and trait prediction. MA-CBxDP establishes new state-of-the-art results in predicting clinical scores and disease status classification, while maintaining exceptional interpretability, verifying its flexibility and versatility in practical applications.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Intelligent Science and Technology, Northwestern Polytechnical University, 127 Youyi Road, 710072 Shaanxi, China
| | - Yan Yang
- Department of Intelligent Science and Technology, Northwestern Polytechnical University, 127 Youyi Road, 710072 Shaanxi, China
| | - Muheng Shang
- Department of Intelligent Science and Technology, Northwestern Polytechnical University, 127 Youyi Road, 710072 Shaanxi, China
| | - Lei Guo
- Department of Intelligent Science and Technology, Northwestern Polytechnical University, 127 Youyi Road, 710072 Shaanxi, China
| | - Daoqiang Zhang
- School of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Jiangning District, 210000 Nanjing, China
| | - Lei Du
- Department of Intelligent Science and Technology, Northwestern Polytechnical University, 127 Youyi Road, 710072 Shaanxi, China
| | | |
Collapse
|
33
|
Tran D, Nguyen H, Pham VD, Nguyen P, Nguyen Luu H, Minh Phan L, Blair DeStefano C, Jim Yeung SC, Nguyen T. A comprehensive review of cancer survival prediction using multi-omics integration and clinical variables. Brief Bioinform 2025; 26:bbaf150. [PMID: 40221959 PMCID: PMC11994034 DOI: 10.1093/bib/bbaf150] [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: 10/27/2024] [Revised: 01/29/2025] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
Cancer is an umbrella term that includes a wide spectrum of disease severity, from those that are malignant, metastatic, and aggressive to benign lesions with very low potential for progression or death. The ability to prognosticate patient outcomes would facilitate management of various malignancies: patients whose cancer is likely to advance quickly would receive necessary treatment that is commensurate with the predicted biology of the disease. Former prognostic models based on clinical variables (age, gender, cancer stage, tumor grade, etc.), though helpful, cannot account for genetic differences, molecular etiology, tumor heterogeneity, and important host biological mechanisms. Therefore, recent prognostic models have shifted toward the integration of complementary information available in both molecular data and clinical variables to better predict patient outcomes: vital status (overall survival), metastasis (metastasis-free survival), and recurrence (progression-free survival). In this article, we review 20 survival prediction approaches that integrate multi-omics and clinical data to predict patient outcomes. We discuss their strategies for modeling survival time (continuous and discrete), the incorporation of molecular measurements and clinical variables into risk models (clinical and multi-omics data), how to cope with censored patient records, the effectiveness of data integration techniques, prediction methodologies, model validation, and assessment metrics. The goal is to inform life scientists of available resources, and to provide a complete review of important building blocks in survival prediction. At the same time, we thoroughly describe the pros and cons of each methodology, and discuss in depth the outstanding challenges that need to be addressed in future method development.
Collapse
Affiliation(s)
- Dao Tran
- Department of Computer Science and Software Engineering, Auburn University, 345 W Magnolia Avenue, Auburn, AL 36849, United States
| | - Ha Nguyen
- Department of Computer Science and Software Engineering, Auburn University, 345 W Magnolia Avenue, Auburn, AL 36849, United States
| | - Van-Dung Pham
- Department of Computer Science and Software Engineering, Auburn University, 345 W Magnolia Avenue, Auburn, AL 36849, United States
| | - Phuong Nguyen
- Department of Computer Science and Software Engineering, Auburn University, 345 W Magnolia Avenue, Auburn, AL 36849, United States
| | - Hung Nguyen Luu
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, 5150 Centre Avenue, Pittsburgh, PA 15232, United States
- Department of Epidemiology, School of Public Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA 15261, United States
| | - Liem Minh Phan
- David Grant USAF Medical Center—Clinical Investigation Facility, 60 Medical Group, Defense Health Agency, 101 Bodin Circle, Travis Air Force Base, CA 94535, United States
| | - Christin Blair DeStefano
- Walter Reed National Military Medical Center, Defense Health Agency, 8901 Rockville Pike, Bethesda, MD 20889, United States
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX 77030, United States
| | - Tin Nguyen
- Department of Computer Science and Software Engineering, Auburn University, 345 W Magnolia Avenue, Auburn, AL 36849, United States
| |
Collapse
|
34
|
Spooner A, Moridani MK, Toplis B, Behary J, Safarchi A, Maher S, Vafaee F, Zekry A, Sowmya A. Benchmarking ensemble machine learning algorithms for multi-class, multi-omics data integration in clinical outcome prediction. Brief Bioinform 2025; 26:bbaf116. [PMID: 40116658 PMCID: PMC11926982 DOI: 10.1093/bib/bbaf116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/06/2025] [Accepted: 02/21/2025] [Indexed: 03/23/2025] Open
Abstract
The complementary information found in different modalities of patient data can aid in more accurate modelling of a patient's disease state and a better understanding of the underlying biological processes of a disease. However, the analysis of multi-modal, multi-omics data presents many challenges. In this work, we compare the performance of a variety of ensemble machine learning (ML) algorithms that are capable of late integration of multi-class data from different modalities. The ensemble methods and their variations tested were (i) a voting ensemble, with hard and soft vote, (ii) a meta learner, and (iii) a multi-modal AdaBoost model using hard vote, soft vote, and meta learner to integrate the modalities on each boosting round, the PB-MVBoost model and a novel application of a mixture of expert's model. These were compared to simple concatenation. We examine these methods using data from an in-house study on hepatocellular carcinoma, plus validation datasets on studies from breast cancer and irritable bowel disease. We develop models that achieve an area under the receiver operating curve of up to 0.85 and find that two boosted methods, PB-MVBoost and AdaBoost with soft vote were the best performing models. We also examine the stability of features selected and the size of the clinical signature. Our work shows that integrating complementary omics and data modalities with effective ensemble ML models enhances accuracy in multi-class clinical outcome predictions and produces more stable predictive features than individual modalities or simple concatenation. We provide recommendations for the integration of multi-modal multi-class data.
Collapse
Affiliation(s)
- Annette Spooner
- School of Computer Science and Engineering, University of New South Wales, High St, Kensington, NSW 2052, Australia
| | - Mohammad Karimi Moridani
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, NSW 2052, Australia
| | - Barbra Toplis
- St George and Sutherland Clinical Campuses, University of New South Wales, Short St, Kogarah, NSW 2217, Australia
| | - Jason Behary
- St George and Sutherland Clinical Campuses, University of New South Wales, Short St, Kogarah, NSW 2217, Australia
- Department of Gastroenterology and Hepatology, St George Hospital, Gray St, Kogarah, NSW 2217, Australia
| | - Azadeh Safarchi
- Health and Biosecurity, Microbiome for One System Health, Commonwealth Scientific and Industrial Research Organisation, 160 Hawkesbury Rd, Westmead, NSW 2145, Australia
| | - Salim Maher
- St George and Sutherland Clinical Campuses, University of New South Wales, Short St, Kogarah, NSW 2217, Australia
- Department of Gastroenterology and Hepatology, St George Hospital, Gray St, Kogarah, NSW 2217, Australia
| | - Fatemeh Vafaee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, NSW 2052, Australia
- UNSW Data Science Hub, University of New South Wales, High St, Kensington, NSW 2052, Australia
| | - Amany Zekry
- St George and Sutherland Clinical Campuses, University of New South Wales, Short St, Kogarah, NSW 2217, Australia
- Department of Gastroenterology and Hepatology, St George Hospital, Gray St, Kogarah, NSW 2217, Australia
| | - Arcot Sowmya
- School of Computer Science and Engineering, University of New South Wales, High St, Kensington, NSW 2052, Australia
| |
Collapse
|
35
|
Boehm KM, El Nahhas OSM, Marra A, Waters M, Jee J, Braunstein L, Schultz N, Selenica P, Wen HY, Weigelt B, Paul ED, Cekan P, Erber R, Loeffler CML, Guerini-Rocco E, Fusco N, Frascarelli C, Mane E, Munzone E, Dellapasqua S, Zagami P, Curigliano G, Razavi P, Reis-Filho JS, Pareja F, Chandarlapaty S, Shah SP, Kather JN. Multimodal histopathologic models stratify hormone receptor-positive early breast cancer. Nat Commun 2025; 16:2106. [PMID: 40025017 PMCID: PMC11873197 DOI: 10.1038/s41467-025-57283-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 02/13/2025] [Indexed: 03/04/2025] Open
Abstract
The Oncotype DX® Recurrence Score (RS) is an assay for hormone receptor-positive early breast cancer with extensively validated predictive and prognostic value. However, its cost and lag time have limited global adoption, and previous attempts to estimate it using clinicopathologic variables have had limited success. To address this, we assembled 6172 cases across three institutions and developed Orpheus, a multimodal deep learning tool to infer the RS from H&E whole-slide images. Our model identifies TAILORx high-risk cases (RS > 25) with an area under the curve (AUC) of 0.89, compared to a leading clinicopathologic nomogram with 0.73. Furthermore, in patients with RS ≤ 25, Orpheus ascertains risk of metastatic recurrence more accurately than the RS itself (0.75 vs 0.49 mean time-dependent AUC). These findings have the potential to guide adjuvant therapy for high-risk cases and tailor surveillance for patients at elevated metastatic recurrence risk.
Collapse
Affiliation(s)
- Kevin M Boehm
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, 323 E 61 St, New York, NY, USA
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Omar S M El Nahhas
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany
- StratifAI GmbH, Suite 14500 Großenhainer Str. 98, 01127, Dresden, Germany
| | - Antonio Marra
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
| | - Michele Waters
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, 323 E 61 St, New York, NY, USA
| | - Justin Jee
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, 323 E 61 St, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Lior Braunstein
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Nikolaus Schultz
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, 323 E 61 St, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Pier Selenica
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Hannah Y Wen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Evan D Paul
- MultiplexDX, s.r.o., Ilkovičova 8, 841 04 Karlova Ves, Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court Suite 450, Rockville, MD, 20850, USA
| | - Pavol Cekan
- MultiplexDX, s.r.o., Ilkovičova 8, 841 04 Karlova Ves, Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc., One Research Court Suite 450, Rockville, MD, 20850, USA
| | - Ramona Erber
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Krankenhausstraße 8-10, 91054, Erlangen, Germany
| | - Chiara M L Loeffler
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Elena Guerini-Rocco
- Department of Pathology, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Nicola Fusco
- Department of Pathology, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Chiara Frascarelli
- Department of Pathology, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Eltjona Mane
- Department of Pathology, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
| | - Elisabetta Munzone
- Division of Medical Senology, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
| | - Silvia Dellapasqua
- Division of Medical Senology, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
| | - Paola Zagami
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Giuseppe Curigliano
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Pedram Razavi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Jorge S Reis-Filho
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
- AstraZeneca, 1 MedImmune Way, Gaithersburg, MD, 20878, USA
| | - Fresia Pareja
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Sarat Chandarlapaty
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA.
| | - Sohrab P Shah
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, 323 E 61 St, New York, NY, USA.
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Im Neuenheimer Feld 460, 69120, Heidelberg, Germany.
| |
Collapse
|
36
|
Yoo SK, Fitzgerald CW, Cho BA, Fitzgerald BG, Han C, Koh ES, Pandey A, Sfreddo H, Crowley F, Korostin MR, Debnath N, Leyfman Y, Valero C, Lee M, Vos JL, Lee AS, Zhao K, Lam S, Olumuyide E, Kuo F, Wilson EA, Hamon P, Hennequin C, Saffern M, Vuong L, Hakimi AA, Brown B, Merad M, Gnjatic S, Bhardwaj N, Galsky MD, Schadt EE, Samstein RM, Marron TU, Gönen M, Morris LGT, Chowell D. Prediction of checkpoint inhibitor immunotherapy efficacy for cancer using routine blood tests and clinical data. Nat Med 2025; 31:869-880. [PMID: 39762425 PMCID: PMC11922749 DOI: 10.1038/s41591-024-03398-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 11/01/2024] [Indexed: 01/25/2025]
Abstract
Predicting whether a patient with cancer will benefit from immune checkpoint inhibitors (ICIs) without resorting to advanced genomic or immunologic assays is an important clinical need. To address this, we developed and evaluated SCORPIO, a machine learning system that utilizes routine blood tests (complete blood count and comprehensive metabolic profile) alongside clinical characteristics from 9,745 ICI-treated patients across 21 cancer types. SCORPIO was trained on data from 1,628 patients across 17 cancer types from Memorial Sloan Kettering Cancer Center. In two internal test sets comprising 2,511 patients across 19 cancer types, SCORPIO achieved median time-dependent area under the receiver operating characteristic curve (AUC(t)) values of 0.763 and 0.759 for predicting overall survival at 6, 12, 18, 24 and 30 months, outperforming tumor mutational burden (TMB), which showed median AUC(t) values of 0.503 and 0.543. Additionally, SCORPIO demonstrated superior predictive performance for predicting clinical benefit (tumor response or prolonged stability), with AUC values of 0.714 and 0.641, compared to TMB (AUC = 0.546 and 0.573). External validation was performed using 10 global phase 3 trials (4,447 patients across 6 cancer types) and a real-world cohort from the Mount Sinai Health System (1,159 patients across 18 cancer types). In these external cohorts, SCORPIO maintained robust performance in predicting ICI outcomes, surpassing programmed death-ligand 1 immunostaining. These findings underscore SCORPIO's reliability and adaptability, highlighting its potential to predict patient outcomes with ICI therapy across diverse cancer types and healthcare settings.
Collapse
Affiliation(s)
- Seong-Keun Yoo
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Conall W Fitzgerald
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Byuri Angela Cho
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bailey G Fitzgerald
- Department of Medicine, Thoracic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Catherine Han
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth S Koh
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Abhinav Pandey
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hannah Sfreddo
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fionnuala Crowley
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brookdale Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Neha Debnath
- Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside and West, New York, NY, USA
| | - Yan Leyfman
- Internal Medicine, Icahn School of Medicine at Mount Sinai South Nassau, Rockville Centre, NY, USA
| | - Cristina Valero
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark Lee
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joris L Vos
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew Sangho Lee
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karena Zhao
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stanley Lam
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ezekiel Olumuyide
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fengshen Kuo
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eric A Wilson
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pauline Hamon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Clotilde Hennequin
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Saffern
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lynda Vuong
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Ari Hakimi
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brian Brown
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nina Bhardwaj
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Galsky
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Robert M Samstein
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas U Marron
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Early Phase Trials Unit, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luc G T Morris
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Diego Chowell
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
37
|
Chang TG, Park S, Schäffer AA, Jiang P, Ruppin E. Hallmarks of artificial intelligence contributions to precision oncology. NATURE CANCER 2025; 6:417-431. [PMID: 40055572 PMCID: PMC11957836 DOI: 10.1038/s43018-025-00917-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/21/2025] [Indexed: 03/29/2025]
Abstract
The integration of artificial intelligence (AI) into oncology promises to revolutionize cancer care. In this Review, we discuss ten AI hallmarks in precision oncology, organized into three groups: (1) cancer prevention and diagnosis, encompassing cancer screening, detection and profiling; (2) optimizing current treatments, including patient outcome prediction, treatment planning and monitoring, clinical trial design and matching, and developing response biomarkers; and (3) advancing new treatments by identifying treatment combinations, discovering cancer vulnerabilities and designing drugs. We also survey AI applications in interventional clinical trials and address key challenges to broader clinical adoption of AI: data quality and quantity, model accuracy, clinical relevance and patient benefit, proposing actionable solutions for each.
Collapse
Affiliation(s)
- Tian-Gen Chang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Seongyong Park
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alejandro A Schäffer
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peng Jiang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
38
|
Momoli C, Costa B, Lenti L, Tubertini M, Parenti MD, Martella E, Varchi G, Ferroni C. The Evolution of Anticancer 3D In Vitro Models: The Potential Role of Machine Learning and AI in the Next Generation of Animal-Free Experiments. Cancers (Basel) 2025; 17:700. [PMID: 40002293 PMCID: PMC11853635 DOI: 10.3390/cancers17040700] [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: 12/20/2024] [Revised: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
The development of anticancer therapies has increasingly relied on advanced 3D in vitro models, which more accurately mimic the tumor microenvironment compared to traditional 2D cultures. This review describes the evolution of these 3D models, highlighting significant advancements and their impact on cancer research. We discuss the integration of machine learning (ML) and artificial intelligence (AI) in enhancing the predictive power and efficiency of these models, potentially reducing the dependence on animal testing. ML and AI offer innovative approaches for analyzing complex data, optimizing experimental conditions, and predicting therapeutic outcomes with higher accuracy. By leveraging these technologies, the next generation of 3D in vitro models could revolutionize anticancer drug development, offering effective alternatives to animal experiments.
Collapse
Affiliation(s)
| | | | | | | | | | - Elisa Martella
- Institute for the Organic Synthesis and Photoreactivity—Italian National Research Council, 40129 Bologna, Italy; (C.M.); (B.C.); (L.L.); (M.T.); (M.D.P.); (C.F.)
| | - Greta Varchi
- Institute for the Organic Synthesis and Photoreactivity—Italian National Research Council, 40129 Bologna, Italy; (C.M.); (B.C.); (L.L.); (M.T.); (M.D.P.); (C.F.)
| | | |
Collapse
|
39
|
Zhang H, Yang F, Xu Y, Zhao S, Jiang YZ, Shao ZM, Xiao Y. Multimodal integration using a machine learning approach facilitates risk stratification in HR+/HER2- breast cancer. Cell Rep Med 2025; 6:101924. [PMID: 39848244 PMCID: PMC11866502 DOI: 10.1016/j.xcrm.2024.101924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 08/11/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025]
Abstract
Hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) breast cancer is the most common type of breast cancer, with continuous recurrence remaining an important clinical issue. Current relapse predictive models in HR+/HER2- breast cancer patients still have limitations. The integration of multidimensional data represents a promising alternative for predicting relapse. In this study, we leverage our multi-omics cohort comprising 579 HR+/HER2- breast cancer patients (200 patients with complete data across 7 modalities) and develop a machine-learning-based model, namely CIMPTGV, which integrates clinical information, immunohistochemistry, metabolomics, pathomics, transcriptomics, genomics, and copy number variations to predict recurrence risk of HR+/HER2- breast cancer. This model achieves concordance indices (C-indices) of 0.871 and 0.869 in the train and test sets, respectively. The risk population predicted by the CIMPTGV model encompasses those identified by single-modality models. Feature analysis reveals that synergistic and complementary effects exist in different modalities. Simultaneously, we develop a simplified model with a mean area under the curve (AUC) of 0.840, presenting a useful approach for clinical applications.
Collapse
Affiliation(s)
- Hang Zhang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China
| | - Fan Yang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China
| | - Ying Xu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China
| | - Shen Zhao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China.
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China.
| |
Collapse
|
40
|
Nittas V, Ormond KE, Vayena E, Blasimme A. Realizing the promise of machine learning in precision oncology: expert perspectives on opportunities and challenges. BMC Cancer 2025; 25:276. [PMID: 39962436 PMCID: PMC11834663 DOI: 10.1186/s12885-025-13621-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 01/31/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND The ability of machine learning (ML) to process and learn from large quantities of heterogeneous patient data is gaining attention in the precision oncology community. Some remarkable developments have taken place in the domain of image classification tasks in areas such as digital pathology and diagnostic radiology. The application of ML approaches to the analysis of DNA data, including tumor-derived genomic profiles, microRNAs, and cancer epigenetic signatures, while relatively more recent, has demonstrated some utility in identifying driver variants and molecular signatures with possible prognostic and therapeutic applications. METHODS We conducted semi-structured interviews with academic and clinical experts to capture the status quo, challenges, opportunities, ethical implications, and future directions. RESULTS Our participants agreed that machine learning in precision oncology is in infant stages, with clinical integration still rare. Overall, participants equated ongoing developments with better clinical workflows and improved treatment decisions for more cancer patients. They underscored the ability of machine learning to tackle the dynamic nature of cancer, break down the complexity of molecular data, and support decision-making. Our participants emphasized obstacles related to molecular data access, clinical utility, and guidelines. The availability of reliable and well-curated data to train and validate machine learning algorithms and integrate multiple data sources were described as constraints yet necessary for future clinical implementation. Frequently mentioned ethical challenges included privacy risks, equity, explainability, trust, and incidental findings, with privacy being the most polarizing. While participants recognized the issue of hype surrounding machine learning in precision oncology, they agreed that, in an assistive role, it represents the future of precision oncology. CONCLUSIONS Given the unique nature of medical AI, our findings highlight the field's potential and remaining challenges. ML will continue to advance cancer research and provide opportunities for patient-centric, personalized, and efficient precision oncology. Yet, the field must move beyond hype and toward concrete efforts to overcome key obstacles, such as ensuring access to molecular data, establishing clinical utility, developing guidelines and regulations, and meaningfully addressing ethical challenges.
Collapse
Affiliation(s)
- Vasileios Nittas
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Hirschengraben 84, Zurich, 8001, Switzerland
- Health Ethics and Policy Lab, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zurich), Hottingerstrasse 10, Zurich, 8092, Switzerland
| | - Kelly E Ormond
- Health Ethics and Policy Lab, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zurich), Hottingerstrasse 10, Zurich, 8092, Switzerland
| | - Effy Vayena
- Health Ethics and Policy Lab, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zurich), Hottingerstrasse 10, Zurich, 8092, Switzerland
| | - Alessandro Blasimme
- Health Ethics and Policy Lab, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zurich), Hottingerstrasse 10, Zurich, 8092, Switzerland.
| |
Collapse
|
41
|
Xu Y, Jiang X, Hu Z. Synergizing metabolomics and artificial intelligence for advancing precision oncology. Trends Mol Med 2025:S1471-4914(25)00016-4. [PMID: 39956738 DOI: 10.1016/j.molmed.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/18/2025]
Abstract
Metabolomics has emerged as a transformative tool in precision oncology, with substantial potential for advancing biomarker discovery, monitoring treatment responses, and aiding drug development. Integrating artificial intelligence (AI) into metabolomics optimizes data acquisition and analysis, facilitating the interpretation of complex metabolic networks and enabling more effective multiomics integration. In this opinion, we explore recent advances in the application of metabolomics within precision oncology, emphasizing the unique advantages that AI-driven metabolomics offers. We propose that AI not only complements but also amplifies the potential of current platforms, accelerating research progress and ultimately improving patient outcomes. Finally, we discuss the opportunities and challenges involved in translating AI-driven metabolomics into clinical practice for precision oncology.
Collapse
Affiliation(s)
- Yipeng Xu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xiaojuan Jiang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
42
|
Banerjee P, Ray S, Dai L, Sandford E, Chatterjee T, Mandal S, Siddiqui J, Tewari M, Walter NG. Chromato-kinetic fingerprinting enables multiomic digital counting of single disease biomarker molecules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.636009. [PMID: 39975368 PMCID: PMC11838488 DOI: 10.1101/2025.01.31.636009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Early and personalized intervention in complex diseases requires robust molecular diagnostics, yet the simultaneous detection of diverse biomarkers-microRNAs (miRNAs), mutant DNAs, and proteins-remains challenging due to low abundance and preprocessing incompatibilities. We present Biomarker Single-molecule Chromato-kinetic multi-Omics Profiling and Enumeration (Bio-SCOPE), a next-generation, triple-modality, multiplexed detection platform that integrates both chromatic and kinetic fingerprinting for molecular profiling through digital encoding. Bio-SCOPE achieves femtomolar sensitivity, single-base mismatch specificity, and minimal matrix interference, enabling precise, parallel quantification of up to six biomarkers in a single sample with single-molecule resolution. We demonstrate its versatility in accurately detecting low-abundance miRNA signatures from human tissues, identifying upregulated miRNAs in the plasma of prostate cancer patients, and measuring elevated interleukin-6 (IL-6) and hsa-miR-21 levels in cytokine release syndrome patients. By seamlessly integrating multiomic biomarker panels on a unified, high-precision platform, Bio-SCOPE provides a transformative tool for molecular diagnostics and precision medicine.
Collapse
Affiliation(s)
- Pavel Banerjee
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Sujay Ray
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Liuhan Dai
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Erin Sandford
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | | | - Shankar Mandal
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Javed Siddiqui
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Muneesh Tewari
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
- Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Nils G. Walter
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
- Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
43
|
Zhou H, Zheng Z, Fan C, Zhou Z. Mechanisms and strategies of immunosenescence effects on non-small cell lung cancer (NSCLC) treatment: A comprehensive analysis and future directions. Semin Cancer Biol 2025; 109:44-66. [PMID: 39793777 DOI: 10.1016/j.semcancer.2025.01.001] [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: 09/20/2024] [Revised: 12/29/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Non-small cell lung cancer (NSCLC), the most prevalent form of lung cancer, remains a leading cause of cancer-related mortality worldwide, particularly among elderly individuals. The phenomenon of immunosenescence, characterized by the progressive decline in immune cell functionality with aging, plays a pivotal role in NSCLC progression and contributes to the diminished efficacy of therapeutic interventions in older patients. Immunosenescence manifests through impaired immune surveillance, reduced cytotoxic responses, and increased chronic inflammation, collectively fostering a pro-tumorigenic microenvironment. This review provides a comprehensive analysis of the molecular, cellular, and genetic mechanisms of immunosenescence and its impact on immune surveillance and the tumor microenvironment (TME) in NSCLC. We explore how aging affects various immune cells, including T cells, B cells, NK cells, and macrophages, and how these changes compromise the immune system's ability to detect and eliminate tumor cells. Furthermore, we address the challenges posed by immunosenescence to current therapeutic strategies, particularly immunotherapy, which faces significant hurdles in elderly patients due to immune dysfunction. The review highlights emerging technologies, such as single-cell sequencing and CRISPR-Cas9, which offer new insights into immunosenescence and its potential as a therapeutic target. Finally, we outline future research directions, including strategies for rejuvenating the aging immune system and optimizing immunotherapy for older NSCLC patients, with the goal of improving treatment efficacy and survival outcomes. These efforts hold promise for the development of more effective, personalized therapies for elderly patients with NSCLC.
Collapse
Affiliation(s)
- Huatao Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Zilong Zheng
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| | - Zijing Zhou
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| |
Collapse
|
44
|
Xiang J, Wang X, Zhang X, Xi Y, Eweje F, Chen Y, Li Y, Bergstrom C, Gopaulchan M, Kim T, Yu KH, Willens S, Olguin FM, Nirschl JJ, Neal J, Diehn M, Yang S, Li R. A vision-language foundation model for precision oncology. Nature 2025; 638:769-778. [PMID: 39779851 DOI: 10.1038/s41586-024-08378-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/08/2024] [Indexed: 01/11/2025]
Abstract
Clinical decision-making is driven by multimodal data, including clinical notes and pathological characteristics. Artificial intelligence approaches that can effectively integrate multimodal data hold significant promise in advancing clinical care1,2. However, the scarcity of well-annotated multimodal datasets in clinical settings has hindered the development of useful models. In this study, we developed the Multimodal transformer with Unified maSKed modeling (MUSK), a vision-language foundation model designed to leverage large-scale, unlabelled, unpaired image and text data. MUSK was pretrained on 50 million pathology images from 11,577 patients and one billion pathology-related text tokens using unified masked modelling. It was further pretrained on one million pathology image-text pairs to efficiently align the vision and language features. With minimal or no further training, MUSK was tested in a wide range of applications and demonstrated superior performance across 23 patch-level and slide-level benchmarks, including image-to-text and text-to-image retrieval, visual question answering, image classification and molecular biomarker prediction. Furthermore, MUSK showed strong performance in outcome prediction, including melanoma relapse prediction, pan-cancer prognosis prediction and immunotherapy response prediction in lung and gastro-oesophageal cancers. MUSK effectively combined complementary information from pathology images and clinical reports and could potentially improve diagnosis and precision in cancer therapy.
Collapse
Affiliation(s)
- Jinxi Xiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiyue Wang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaoming Zhang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yinghua Xi
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Feyisope Eweje
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yijiang Chen
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuchen Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Colin Bergstrom
- Department of Medicine (Oncology), Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew Gopaulchan
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ted Kim
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Sierra Willens
- Department of Medicine (Oncology), Stanford University School of Medicine, Stanford, CA, USA
| | - Francesca Maria Olguin
- Department of Medicine (Oncology), Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey J Nirschl
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joel Neal
- Department of Medicine (Oncology), Stanford University School of Medicine, Stanford, CA, USA
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sen Yang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Institute for Human-Centered Artificial Intelligence, Stanford, CA, USA.
| |
Collapse
|
45
|
Ben Cohen G, Yaacov A, Ben Zvi Y, Loutati R, Lishinsky N, Landau J, Hope T, Popovzter A, Rosenberg S. Graph convolution networks model identifies and quantifies gene and cancer specific transcriptome signatures of cancer driver events. Comput Biol Med 2025; 185:109491. [PMID: 39700860 DOI: 10.1016/j.compbiomed.2024.109491] [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: 05/20/2024] [Revised: 08/24/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND The identification and drug targeting of cancer causing (driver) genetic alterations has seen immense improvement in recent years, with many new targeted therapies developed. However, identifying, prioritizing, and treating genetic alterations is insufficient for most cancer patients. Current clinical practices rely mainly on DNA level mutational analyses, which in many cases fail to identify treatable driver events. Arguably, signal strength may determine cell fate more than the mutational status that initiated it. The use of transcriptomics, a complex and highly informative representation of cellular and tumor state, had been suggested to enhance diagnostics and treatment successes. A gene-expression based model trained over known genetic alterations could improve identification and quantification of cancer related biological aberrations' signal strength. METHODS We present STAMP (Signatures in Transcriptome Associated with Mutated Protein), a Graph Convolution Networks (GCN) based framework for the identification of gene expression signatures related to cancer driver events. STAMP was trained to identify the p53 dysfunction of cancer samples from gene expression, utilizing comprehensive curated graph structures of gene interactions. Predictions were modified for generating a quantitative score to rank the severity of a driver event in each sample. STAMP was then extended to almost 300 tumor type-specific predictive models for important cancer genes/pathways, by training to identify well-established driver events' annotations from the literature. RESULTS STAMP achieved very high AUC on unseen data across several tumor types and on an independent cohort. The framework was validated on p53 related genetic and clinical characteristics, including the effect of Variants of Unknown Significance, and showed strong correlation with protein function. For genes and tumor types where targeted therapy is available, STAMP showed correlation with drugs sensitivity (IC50) in an independent cell line database. It managed to stratify drug effect on samples with similar mutational profiles. STAMP was validated for drug-response prediction in clinical patients' cohorts, improving over a state-of-the-art method and suggesting potential biomarkers for cancer treatments. CONCLUSIONS The STAMP models provide a learning framework that successfully identifies and quantifies driver events' signal strength, showing utility in portraying the molecular landscape of tumors based on transcriptomics. Importantly, STAMP manifested the ability to improve targeted therapy selection and hence can contribute to better treatment.
Collapse
Affiliation(s)
- Gil Ben Cohen
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| | - Adar Yaacov
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Yishai Ben Zvi
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Ranel Loutati
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Natan Lishinsky
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Jakob Landau
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Tom Hope
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Israel
| | - Aron Popovzter
- Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Shai Rosenberg
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| |
Collapse
|
46
|
Martínez-Ramírez JM, Carmona C, Ramírez-Expósito MJ, Martínez-Martos JM. Extracting Knowledge from Machine Learning Models to Diagnose Breast Cancer. Life (Basel) 2025; 15:211. [PMID: 40003620 PMCID: PMC11856414 DOI: 10.3390/life15020211] [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: 12/31/2024] [Revised: 01/17/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
This study explored the application of explainable machine learning models to enhance breast cancer diagnosis using serum biomarkers, contrary to many studies that focus on medical images and demographic data. The primary objective was to develop models that are not only accurate but also provide insights into the factors driving predictions, addressing the need for trustworthy AI in healthcare. Several classification models were evaluated, including OneR, JRIP, the FURIA, J48, the ADTree, and the Random Forest, all of which are known for their explainability. The dataset included a variety of biomarkers, such as electrolytes, metal ions, marker proteins, enzymes, lipid profiles, peptide hormones, steroid hormones, and hormone receptors. The Random Forest model achieved the highest accuracy at 99.401%, followed closely by JRIP, the FURIA, and the ADTree at 98.802%. OneR and J48 achieved 98.204% accuracy. Notably, the models identified oxytocin as a key predictive biomarker, with most models featuring it in their rules. Other significant parameters included GnRH, β-endorphin, vasopressin, IRAP, and APB, as well as factors like iron, cholinesterase, the total protein, progesterone, 5-nucleotidase, and the BMI, which are considered clinically relevant to breast cancer pathogenesis. This study discusses the roles of the identified parameters in cancer development, thus underscoring the potential of explainable machine learning models for enhancing early breast cancer diagnosis by focusing on explainability and the use of serum biomarkers.The combination of both can lead to improved early detection and personalized treatments, emphasizing the potential of these methods in clinical settings. The identified markers also provide additional research and therapeutic targets for breast cancer pathogenesis and a deep understanding of their interactions, advancing personalized approaches to breast cancer management.
Collapse
Affiliation(s)
| | - Cristobal Carmona
- Department of Computer Science, University of Jaén, E-23071 Jaén, Spain; (J.M.M.-R.); (C.C.)
- Andalusian Research Institute in Data Science and Computational Intelligence, DASCI, University of Jaén, E-23071 Jaén, Spain
- Leicester School of Pharmacy, DeMontfort University, Leicester LE1 7RH, UK
| | - María Jesús Ramírez-Expósito
- Experimental and Clinical Physiopathology Research Group CVI-1039, Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain;
| | - José Manuel Martínez-Martos
- Experimental and Clinical Physiopathology Research Group CVI-1039, Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain;
| |
Collapse
|
47
|
van Wilpe S, Croci D, Fonseca Costa SS, te Paske IB, Tolmeijer SH, van Ipenburg J, Kroeze LI, Pavan S, Monnier-Benoit S, Coccia G, Hadadi N, Oving IM, Smilde TJ, van Voorthuizen T, Berends M, Franken MD, Ligtenberg MJ, Hosseinian Ehrensberger S, Ciarloni L, Romero P, Mehra N. Multimodal integration of blood RNA and ctDNA reflects response to immunotherapy in metastatic urothelial cancer. JCI Insight 2025; 10:e186062. [PMID: 39883530 PMCID: PMC11949011 DOI: 10.1172/jci.insight.186062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Previously, we demonstrated that changes in circulating tumor DNA (ctDNA) are promising biomarkers for early response prediction (ERP) to immune checkpoint inhibitors (ICIs) in metastatic urothelial cancer (mUC). In this study, we investigated the value of whole-blood immunotranscriptomics for ERP-ICI and integrated both biomarkers into a multimodal model to boost accuracy. METHODS Blood samples of 93 patients were collected at baseline and after 2-6 weeks of ICI for ctDNA (n = 88) and immunotranscriptome (n = 79) analyses. ctDNA changes were dichotomized into increase or no increase, the latter including patients with undetectable ctDNA. For RNA model development, the cohort was split into discovery (n = 29), test (n = 29), and validation sets (n = 21). Finally, RNA- and ctDNA-based predictions were integrated in a multimodal model. Clinical benefit (CB) was defined as progression-free survival beyond 6 months. RESULTS Sensitivity (SN) and specificity (SP) of ctDNA increase for predicting non-CB (N-CB) was 59% and 92%, respectively. Immunotranscriptome analysis revealed upregulation of T cell activation, proliferation, and interferon signaling during treatment in the CB group, in contrast with N-CB patients. Based on these differences, a 10-gene RNA model was generated, reaching an SN and SP of 73% and 79%, respectively, in the test and 67% and 67% in the validation set for predicting N-CB. Multimodal model integration led to superior performance, with an SN and SP of 79% and 100%, respectively, in the validation cohort. CONCLUSION The combination of whole-blood immunotranscriptome and ctDNA in a multimodal model showed promise for ERP-ICI in mUC and accurately identified patients with N-CB. FUNDING Eurostars grant E! 114908 - PRECISE, Paul Speth Foundation (Bullseye project).
Collapse
Affiliation(s)
- Sandra van Wilpe
- Medical Oncology Department, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | | | | | - Iris B.A.W. te Paske
- Medical Oncology Department, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Sofie H. Tolmeijer
- Medical Oncology Department, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jolique van Ipenburg
- Department of Pathology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Leonie I. Kroeze
- Department of Pathology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | | | | | | | | | - Irma M. Oving
- Department of Medical Oncology, Ziekenhuisgroep Twente, Almelo, Netherlands
| | - Tineke J. Smilde
- Department of Medical Oncology, Jeroen Bosch Ziekenhuis, ‘s-Hertogenbosch, Netherlands
| | | | - Marieke Berends
- Department of Medical Oncology, Canisius Wilhelmina Ziekenhuis, Nijmegen, Netherlands
| | - Mira D. Franken
- Medical Oncology Department, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Marjolijn J.L. Ligtenberg
- Department of Human Genetics, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | | | | | | | - Niven Mehra
- Medical Oncology Department, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
48
|
Wang Q, Zhao F, Zhang H, Chu T, Wang Q, Pan X, Chen Y, Zhou H, Zheng T, Li Z, Lin F, Xie H, Ma H, Liu L, Zhang L, Li Q, Wang W, Dai Y, Tang R, Wang J, Yang P, Mao N. Deep learning-based multi-task prediction of response to neoadjuvant chemotherapy using multiscale whole slide images in breast cancer: A multicenter study. Chin J Cancer Res 2025; 37:28-47. [PMID: 40078559 PMCID: PMC11893347 DOI: 10.21147/j.issn.1000-9604.2025.01.03] [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: 10/10/2024] [Accepted: 12/20/2024] [Indexed: 03/14/2025] Open
Abstract
Objective Early predicting response before neoadjuvant chemotherapy (NAC) is crucial for personalized treatment plans for locally advanced breast cancer patients. We aim to develop a multi-task model using multiscale whole slide images (WSIs) features to predict the response to breast cancer NAC more finely. Methods This work collected 1,670 whole slide images for training and validation sets, internal testing sets, external testing sets, and prospective testing sets of the weakly-supervised deep learning-based multi-task model (DLMM) in predicting treatment response and pCR to NAC. Our approach models two-by-two feature interactions across scales by employing concatenate fusion of single-scale feature representations, and controls the expressiveness of each representation via a gating-based attention mechanism. Results In the retrospective analysis, DLMM exhibited excellent predictive performance for the prediction of treatment response, with area under the receiver operating characteristic curves (AUCs) of 0.869 [95% confidence interval (95% CI): 0.806-0.933] in the internal testing set and 0.841 (95% CI: 0.814-0.867) in the external testing sets. For the pCR prediction task, DLMM reached AUCs of 0.865 (95% CI: 0.763-0.964) in the internal testing and 0.821 (95% CI: 0.763-0.878) in the pooled external testing set. In the prospective testing study, DLMM also demonstrated favorable predictive performance, with AUCs of 0.829 (95% CI: 0.754-0.903) and 0.821 (95% CI: 0.692-0.949) in treatment response and pCR prediction, respectively. DLMM significantly outperformed the baseline models in all testing sets (P<0.05). Heatmaps were employed to interpret the decision-making basis of the model. Furthermore, it was discovered that high DLMM scores were associated with immune-related pathways and cells in the microenvironment during biological basis exploration. Conclusions The DLMM represents a valuable tool that aids clinicians in selecting personalized treatment strategies for breast cancer patients.
Collapse
Affiliation(s)
- Qin Wang
- School of Information and Electronic Engineering, Shandong Technology and Business University, Yantai 264005, China
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Feng Zhao
- School of Computer Science and Technology, Shandong Technology and Business University, Yantai 264005, China
| | - Haicheng Zhang
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Tongpeng Chu
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Qi Wang
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Xipeng Pan
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin 541004, China
| | - Yuqian Chen
- School of Information and Electronic Engineering, Shandong Technology and Business University, Yantai 264005, China
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Heng Zhou
- School of Information and Electronic Engineering, Shandong Technology and Business University, Yantai 264005, China
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Tiantian Zheng
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Ziyin Li
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Fan Lin
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Haizhu Xie
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Heng Ma
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Lan Liu
- Department of Radiology, Jiangxi Cancer Hospital, the Second Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China
| | - Lina Zhang
- Department of Radiology, the First Affiliated Hospital of China Medical University, Shenyang 400042, China
| | - Qin Li
- Department of Radiology, Weifang Hospital of Traditional Chinese Medicine, Weifang 262600, China
| | - Weiwei Wang
- Department of Medical Imaging, Affiliated Hospital of Jining Medical University, Jining 272029, China
| | - Yi Dai
- Department of Radiology, the Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Ruijun Tang
- Department of Pathology, Guilin Traditional Chinese Medicine Hospital, Guilin 541002, China
| | - Jigang Wang
- Department of Pathology, the Affiliated Hospital of Qingdao University, Qingdao 266555, China
| | - Ping Yang
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Pathology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Ning Mao
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| |
Collapse
|
49
|
MacDonald WJ, Purcell C, Pinho-Schwermann M, Stubbs NM, Srinivasan PR, El-Deiry WS. Heterogeneity in Cancer. Cancers (Basel) 2025; 17:441. [PMID: 39941808 PMCID: PMC11816170 DOI: 10.3390/cancers17030441] [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: 12/19/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Cancer heterogeneity is a major challenge in oncology, complicating diagnosis, prognostication, and treatment. The clinical heterogeneity of cancer, which leads to differential treatment outcomes between patients with histopathologically similar cancers, is attributable to molecular diversity manifesting through genetic, epigenetic, transcriptomic, microenvironmental, and host biology differences. Heterogeneity is observed between patients, individual metastases, and within individual lesions. This review discusses clinical implications of heterogeneity, emphasizing need for personalized approaches to overcome challenges posed by cancer's diverse presentations. Understanding of emerging molecular diagnostic and analytical techniques can provide a view into the multidimensional complexity of cancer heterogeneity. With over 90% of cancer-related deaths associated with metastasis, we additionally explore the role heterogeneity plays in treatment resistance and recurrence of metastatic lesions. Molecular insights from next-generation sequencing, single-cell transcriptomics, liquid biopsy technology, and artificial intelligence will facilitate the development of combination therapy regimens that can potentially induce lasting and even curative treatment outcomes.
Collapse
Affiliation(s)
- William J. MacDonald
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Connor Purcell
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Maximilian Pinho-Schwermann
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Nolan M. Stubbs
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Praveen R. Srinivasan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- The Joint Program in Cancer Biology, Brown University and Brown University Health, Providence, RI 02903, USA
- Hematology-Oncology Division, Department of Medicine, Rhode Island Hospital, Brown University, Providence, RI 02903, USA
| |
Collapse
|
50
|
Ding C, Liao Q, Zuo R, Zhang S, Guo Z, He J, Ye Z, Chen W, Ke S. Machine learning potential predictor of idiopathic pulmonary fibrosis. Front Genet 2025; 15:1464471. [PMID: 39935693 PMCID: PMC11811625 DOI: 10.3389/fgene.2024.1464471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/26/2024] [Indexed: 02/13/2025] Open
Abstract
Introduction Idiopathic pulmonary fibrosis (IPF) is a severe chronic respiratory disease characterized by treatment challenges and poor prognosis. Identifying relevant biomarkers for effective early-stage risk prediction is therefore of critical importance. Methods In this study, we obtained gene expression profiles and corresponding clinical data of IPF patients from the GEO database. GO enrichment and KEGG pathway analyses were performed using R software. To construct an IPF risk prediction model, we employed LASSO-Cox regression analysis and the SVM-RFE algorithm. PODNL1 and PIGA were identified as potential biomarkers associated with IPF onset, and their predictive accuracy was confirmed using ROC curve analysis in the test set. Furthermore, GSEA revealed enrichment in multiple pathways, while immune function analysis demonstrated a significant correlation between IPF onset and immune cell infiltration. Finally, the roles of PODNL1 and PIGA as biomarkers were validated through in vivo and in vitro experiments using qRT-PCR, Western blotting, and immunohistochemistry. Results These findings suggest that PODNL1 and PIGA may serve as critical biomarkers for IPF onset and contribute to its pathogenesis. Discussion This study highlights their potential for early biomarker discovery and risk prediction in IPF, offering insights into disease mechanisms and diagnostic strategies.
Collapse
Affiliation(s)
- Chenchun Ding
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Quan Liao
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Renjie Zuo
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shichao Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhenzhen Guo
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Junjie He
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Ziwei Ye
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Weibin Chen
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Sunkui Ke
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|