1
|
Norouzi S, Hajizadeh E, Jafarabadi MA, Naderi N, Mazloomzadeh S. Deep neural network base competing risk in predicting heart failure patient's survival. J Diabetes Metab Disord 2025; 24:109. [PMID: 40313916 PMCID: PMC12040772 DOI: 10.1007/s40200-025-01595-8] [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: 12/05/2024] [Accepted: 02/04/2025] [Indexed: 05/03/2025]
Abstract
Objectives Heart failure (HF) is a complicated disease with several competing risks of interest, such as HF death and other causes. This study compares a Deep Neural Network Competing Risks (DNNCR) and Random Survival Forest (RSF) model to evaluate the predictive performance of time-to-event outcomes in HF patients with competing risks. Methods This study represents the retrospective analysis of 435 HF patients admitted to RCMRC, Tehran, Iran, between March and August 2018. After a five-year follow-up in 2023, predictions were analyzed based on Cause of death. This study employed RSF and DNN methods to account for competing risks in survival analysis. Then, model fitness was applied using C-index and IBS. Results The C-index of the results shows that DNNCR is superior to RSF in predicting survival outcomes for HF and other causes of death. Precisely, the C-index was 0.65 (0.04) for HF and 0.63 (0.02) for other causes of death in the DNNCR model, while in RSF, the C-index was 0.65 (0.04) for HF and 0.61 (0.03) for Other Causes. Additionally, calibration results showed via the IBS the finest performance of the DNNCR model at 0.16 for HF, followed by other causes with an IBS of 0.18. Conclusions The study shows that the DNNCR model outperforms RSF in predicting survival outcomes for HF patients, particularly in the presence of competing risks. The improved accuracy enables physicians to identify high-risk individuals and tailor treatment plans accordingly. Future research could utilize more diverse datasets to enhance DNNCR performance and integrate these models into clinical tools. Graphical Abstract
Collapse
Affiliation(s)
- Solmaz Norouzi
- Department of Biostatistics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ebrahim Hajizadeh
- Department of Biostatistics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Asghari Jafarabadi
- Biostatistics Unit, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3004 Australia
- Department of Psychiatry, School of Clinical Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3168 Australia
| | - Nasim Naderi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Saeideh Mazloomzadeh
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Teramachi R, Furukawa T, Kondoh Y, Karasuyama M, Hozumi H, Kataoka K, Oyama S, Suda T, Shiratori Y, Ishii M. Deep Learning for Predicting Acute Exacerbation and Mortality of Interstitial Lung Disease. Ann Am Thorac Soc 2025; 22:689-697. [PMID: 39680875 DOI: 10.1513/annalsats.202403-284oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024] Open
Abstract
Rationale: Some patients with interstitial lung disease (ILD) have a high mortality rate or experience acute exacerbation of ILD (AE-ILD) that results in increased mortality. Early identification of these high-risk patients and accurate prediction of the onset of these important events is important to determine treatment strategies. Although various factors that affect disease behavior among patients with ILD hinder the accurate prediction of these events, the use of longitudinal information may enable better prediction. Objectives: To develop a deep learning (DL) model to predict composite outcomes defined as the first occurrence of AE-ILD and mortality using longitudinal data. Methods: Longitudinal clinical and environmental data were retrospectively collected from consecutive patients with ILD at two specialty centers between January 2008 and December 2015. A DL model was developed to predict composite outcomes using longitudinal data from 80% of patients from the first center, which was then validated using data from the remaining 20% of patients and the second center. The developed model was compared with the univariate Cox proportional hazard (CPH) model using the ILD gender-age-physiology (ILD-GAP) score and multivariate CPH model at the time of ILD diagnosis. Results: AE-ILD was reported in 218 patients among the 1,175 patients enrolled, whereas 380 died without developing AE-ILD. The truncated concordance index (C-index) values of univariate/multivariate CPH models for composite outcomes within 12, 24, and 36 months after prediction were 0.789/0.843, 0.788/0.853, and 0.787/0.853 in internal validation, and 0.650/0.718, 0.652/0.756, and 0.640/0.756 in external validation, respectively. At 12 months after ILD diagnosis, the DL model outperformed the univariate CPH model and multivariate CPH model for composite outcomes within 12 months, with concordance index values of 0.842, 0.840, and 0.839 in internal validation, and 0.803, 0.744, and 0.746 in external validation, respectively. Neutrophils, C-reactive protein, ILD-GAP score, and exposure to suspended particulate matter were strongly associated with the composite outcomes. Conclusions: The DL model can accurately predict the incidence of AE-ILD or mortality using longitudinal data.
Collapse
Affiliation(s)
- Ryo Teramachi
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Taiki Furukawa
- Medical IT Center, Nagoya University Hospital, Nagoya, Japan
| | - Yasuhiro Kondoh
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Japan
| | - Masayuki Karasuyama
- Department of Computer Science, Nagoya Institute of Technology, Nagoya, Japan
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan; and
| | - Kensuke Kataoka
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Japan
| | - Shintaro Oyama
- Center for Preventive Medical Engineering, Nagoya University, Nagoya, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan; and
| | | | - Makoto Ishii
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
3
|
Mao Y, Ding M, Zong D, Mu Z, He X. Predicting radiation-induced hypothyroidism in nasopharyngeal carcinoma patients using a deep learning model. Clin Transl Radiat Oncol 2025; 52:100946. [PMID: 40144227 PMCID: PMC11937289 DOI: 10.1016/j.ctro.2025.100946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/05/2024] [Accepted: 03/09/2025] [Indexed: 03/28/2025] Open
Abstract
Background Radiation-induced hypothyroidism (RIHT) is a common complication in nasopharyngeal carcinoma patients. Predicting its onset is crucial for effective management and early intervention. This study aims to develop a model based on deep learning survival analysis to predict RIHT in nasopharyngeal carcinoma patients. Methods This retrospective study included 535 nasopharyngeal carcinoma patients between January 2015 and October 2020. Cox regression, LASSO-Cox analyses and Spearman correlation test were employed to identify significant predictors. Two deep learning and two machine learning algorithms were trained, tuned, and compared against traditional Cox and NTCP models by C-index, Brier score, and decision curve analysis. Results The study observed a 41.7 % incidence of RIHT, with a median time to onset of 15 months. AJCC N stage, thyroid volume and specific dose-volume parameters were identified as potential predictors. DeepSurv model outperformed traditional ones (C-index: DeepSurv 0.75, traditional models ≤ 0.63). While other models were competitive at early post-treatment intervals, deep learning models demonstrated superior performance over time. Calibration and decision curve analysis corroborated the enhanced predictive capability of DeepSurv. Feature importance analysis highlighted thyroid V30 and V50 as the most significant predictors. Conclusions DeepSurv demonstrated superior predictive performance for RIHT in nasopharyngeal carcinoma patients compared to traditional models. Deep learning-based predictions offer high accuracy, which may enable personalized patient management and have great potentials in mitigating the risk of RIHT. These findings suggested that incorporating such model into clinical practice could be beneficial for the management of RITH.
Collapse
Affiliation(s)
- Yichen Mao
- The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Mingjun Ding
- The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Dan Zong
- The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Zhongde Mu
- The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xia He
- The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Albu E, Gao S, Stijnen P, Rademakers FE, Janssens C, Cossey V, Debaveye Y, Wynants L, Van Calster B. Hospital-wide, dynamic, individualized prediction of central line-associated bloodstream infections-development and temporal evaluation of six prediction models. BMC Infect Dis 2025; 25:597. [PMID: 40275180 PMCID: PMC12023667 DOI: 10.1186/s12879-025-10854-1] [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: 11/05/2024] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Central line-associated bloodstream infections (CLABSI) are preventable hospital-acquired infections. Predicting CLABSI helps improve early intervention strategies and enhance patient safety. AIM To develop and temporally evaluate dynamic prediction models for continuous CLABSI risk monitoring. METHODS Data from hospitalized patients with central catheter(s) admitted to University Hospitals Leuven between 2014 and 2017 were used to develop five dynamic models (a landmark cause-specific model, two random forest models, and two XGBoost models) to predict 7-day CLABSI risk, accounting for competing events (death, discharge, and catheter removal). The models' predictions were then combined using a superlearner model. All models were temporally evaluated on data from the same hospital from 2018 to 2020 using performance metrics for discrimination, calibration, and clinical utility. FINDINGS Among 61629 catheter episodes in the training set, 1930 (3.1%) resulted in CLABSI, while in the test set of 44544 catheter episodes, 1059 (2.4%) experienced CLABSI. Among individual models, one XGBoost model achieved the highest AUROC of 0.748. Calibration was good for predicted risks up to 5%, while the cause-specific and XGBoost models overestimated higher predicted risks. The superlearner displayed a modest improvement in discrimination (AUROC up to 0.751) and better calibration than the cause-specific and XGBoost models, but worse than the random forest models. The models showed clinical utility to support standard care interventions (at risk thresholds between 0.5-4%), but not to support advanced interventions (at thresholds 15-25%). CONCLUSION Hospital-wide CLABSI prediction models offer clinical utility based on medium-risk thresholds. Clinical utility at present may be limited as the model performance deteriorated over time.
Collapse
Affiliation(s)
- Elena Albu
- Department of Development & Regeneration, KU Leuven, Louvain, Belgium
- Leuven Unit for Health Technology Assessment Research (LUHTAR), KU Leuven, Louvain, Belgium
| | - Shan Gao
- Department of Development & Regeneration, KU Leuven, Louvain, Belgium
- Leuven Unit for Health Technology Assessment Research (LUHTAR), KU Leuven, Louvain, Belgium
| | - Pieter Stijnen
- Management Information Reporting Department, University Hospitals Leuven, Louvain, Belgium
| | | | - Christel Janssens
- Vascular Access Specialty Team, University Hospitals Leuven, Louvain, Belgium
| | - Veerle Cossey
- Department of Development & Regeneration, KU Leuven, Louvain, Belgium
- Department of Infection Control and Prevention, University Hospitals Leuven, Louvain, Belgium
| | - Yves Debaveye
- Department of Cellular and Molecular Medicine, University Hospitals Leuven, Louvain, Belgium
| | - Laure Wynants
- Department of Development & Regeneration, KU Leuven, Louvain, Belgium
- Leuven Unit for Health Technology Assessment Research (LUHTAR), KU Leuven, Louvain, Belgium
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
| | - Ben Van Calster
- Department of Development & Regeneration, KU Leuven, Louvain, Belgium.
- Leuven Unit for Health Technology Assessment Research (LUHTAR), KU Leuven, Louvain, Belgium.
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
5
|
Ramakrishnaiah Y, Macesic N, Webb GI, Peleg AY, Tyagi S. EHR-ML: A data-driven framework for designing machine learning applications with electronic health records. Int J Med Inform 2025; 196:105816. [PMID: 39891983 DOI: 10.1016/j.ijmedinf.2025.105816] [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: 12/18/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/03/2025]
Abstract
OBJECTIVE The healthcare landscape is experiencing a transformation with the integration of Artificial Intelligence (AI) into traditional analytic workflows. However, its integration faces challenges resulting in a crisis of generalisability. Key obstacles include; 1) Insufficient consideration of local contextual factors, such as institution-specific data formats, practices, and protocols, which can lead to variability in clinical practices across different institutions. 2) ad-hoc data preparation and design of machine learning strategies. 3) manual subjective adjustment of design parameters resulting in sub-optimal performance. 4) EHR specific challenges regarding data biases affecting the model outcomes and unique intermittent temporal nature of the data necessitating specialised handling 5) lack of cross-institutional data validations. METHODS To address these challenges, EHR-ML, provides an easy to use structured framework for designing optimum machine learning applications in a data-driven manner. The framework supports ingestion of local institutional electronic health records (EHRs) and process standardisation. The study design and parameter optimisation is done in a fully data-driven evidence-based approach. It seamlessly integrating with existing quality control tools. To handle the unique characteristics of the EHR data, it offers customisable ensemble models. It enables the acquisition of EHR data from diverse systems and harmonise them into common formats following international standards. RESULTS The effectiveness of the EHR-ML is demonstrated through a series of case studies. These studies highlight its capability to develop high-performance models in a fully automated manner, consistently surpassing the performance of traditional methodologies. Furthermore, they exhibited strong generalisability across diverse healthcare settings. DISCUSSION AND CONCLUSION EHR-ML enhances the clinical relevance and accuracy of predictive models by incorporating local context into machine learning applications. Additionally, by providing an user-friendly fully-automated framework, it facilitates rapid hypothesis testing aimed to generate localised biomedical knowledge.
Collapse
Affiliation(s)
- Yashpal Ramakrishnaiah
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, 3000, VIC, Australia
| | - Nenad Macesic
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, 3000, VIC, Australia; Centre to Impact AMR, Monash University, Melbourne, 3000, VIC, Australia
| | - Geoffrey I Webb
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, 3000, VIC, Australia; Centre to Impact AMR, Monash University, Melbourne, 3000, VIC, Australia
| | - Anton Y Peleg
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, 3000, VIC, Australia; Centre to Impact AMR, Monash University, Melbourne, 3000, VIC, Australia
| | - Sonika Tyagi
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, 3000, VIC, Australia; School of Computing Technologies, RMIT University, Melbourne, 3000, VIC, Australia.
| |
Collapse
|
6
|
Truchot A, Raynaud M, Helanterä I, Aubert O, Kamar N, Divard G, Astor B, Legendre C, Hertig A, Buchler M, Crespo M, Akalin E, Pujol GS, Ribeiro de Castro MC, Matas AJ, Ulloa C, Jordan SC, Huang E, Juric I, Basic-Jukic N, Coemans M, Naesens M, Friedewald JJ, Silva HT, Lefaucheur C, Segev DL, Collins GS, Loupy A. Competing and Noncompeting Risk Models for Predicting Kidney Allograft Failure. J Am Soc Nephrol 2025; 36:688-701. [PMID: 40168162 PMCID: PMC11975249 DOI: 10.1681/asn.0000000517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024] Open
Abstract
Background Prognostic models are becoming increasingly relevant in clinical trials as potential surrogate end points and for patient management as clinical decision support tools. However, the effect of competing risks on model performance remains poorly investigated. We aimed to carefully assess the performance of competing risk and noncompeting risk models in the context of kidney transplantation, where allograft failure and death with a functioning graft are two competing outcomes. Methods We included 11,046 kidney transplant recipients enrolled in ten countries. We developed prediction models for long-term kidney graft failure prediction, without accounting (i.e., censoring) and accounting for the competing risk of death with a functioning graft, using Cox, Fine–Gray, and cause-specific Cox regression models. To this aim, we followed a detailed and transparent analytical framework for competing and noncompeting risk modeling and carefully assessed the models' development, stability, discrimination, calibration, overall fit, clinical utility, and generalizability in external validation cohorts and subpopulations. More than 15 metrics were used to provide an exhaustive assessment of model performance. Results Among 11,046 recipients in the derivation and validation cohorts, 1497 (14%) lost their graft and 1003 (9%) died with a functioning graft after a median follow-up postrisk evaluation of 4.7 years (interquartile range, 2.7–7.0). The cumulative incidence of graft loss was similarly estimated by Kaplan–Meier and Aalen–Johansen methods (17% versus 16% in the derivation cohort). Cox and competing risk models showed similar and stable risk estimates for predicting long-term graft failure (average mean absolute prediction error of 0.0140, 0.0138, and 0.0135 for Cox, Fine–Gray, and cause-specific Cox models, respectively). Discrimination and overall fit were comparable in the validation cohorts, with concordance index ranging from 0.76 to 0.87. Across various subpopulations and clinical scenarios, the models performed well and similarly, although in some high-risk groups (such as donors older than 65 years), the findings suggest a trend toward moderately improved calibration when using a competing risk approach. Conclusions Competing and noncompeting risk models performed similarly in predicting long-term kidney graft failure.
Collapse
Affiliation(s)
- Agathe Truchot
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| | - Marc Raynaud
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| | - Ilkka Helanterä
- Department of Transplantation and Liver Surgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Olivier Aubert
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
- Kidney Transplant Department, Necker Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Nassim Kamar
- Department of Nephrology and Organ Transplantation, Toulouse Rangueil University Hospital, INSERM UMR 1291, Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University Paul Sabatier, Toulouse, France
| | - Gillian Divard
- Kidney Transplant Department, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Brad Astor
- Division of Nephrology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Christophe Legendre
- Kidney Transplant Department, Necker Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Alexandre Hertig
- Department of Nephrology and Kidney Transplantation, Foch Hospital, Suresnes, France
| | - Matthias Buchler
- Nephrology and Immunology Department, Bretonneau Hospital, Tours, France
| | - Marta Crespo
- Department of Nephrology, Hospital del Mar Barcelona, Barcelona, Spain
| | - Enver Akalin
- Kidney Transplantation Program, Renal Division Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Gervasio Soler Pujol
- Centro de Educacion Medica e Investigaciones Clinicas Buenos Aires, Unidad de Trasplante Renopancreas, Buenos Aires, Argentina
| | | | - Arthur J. Matas
- Division of Transplantation, Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| | | | - Stanley C. Jordan
- Division of Nephrology, Department of Medicine, Comprehensive Transplant Center, Cedars Sinai Medical Center, Los Angeles, California
| | - Edmund Huang
- Division of Nephrology, Department of Medicine, Comprehensive Transplant Center, Cedars Sinai Medical Center, Los Angeles, California
| | - Ivana Juric
- Department of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Center Zagreb, School of Medicine University of Zagreb, Zagreb, Croatia
| | - Nikolina Basic-Jukic
- Department of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Center Zagreb, School of Medicine University of Zagreb, Zagreb, Croatia
| | - Maarten Coemans
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | | | - Helio Tedesco Silva
- Hospital do Rim, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - Carmen Lefaucheur
- Kidney Transplant Department, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Dorry L. Segev
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gary S. Collins
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Centre for Statistics in Medicine, University of Oxford, Oxford, United Kingdom
| | - Alexandre Loupy
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
- Kidney Transplant Department, Necker Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France
| |
Collapse
|
7
|
He R, Sarwal V, Qiu X, Zhuang Y, Zhang L, Liu Y, Chiang J. Generative AI Models in Time-Varying Biomedical Data: Scoping Review. J Med Internet Res 2025; 27:e59792. [PMID: 40063929 PMCID: PMC11933772 DOI: 10.2196/59792] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/08/2024] [Accepted: 11/15/2024] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Trajectory modeling is a long-standing challenge in the application of computational methods to health care. In the age of big data, traditional statistical and machine learning methods do not achieve satisfactory results as they often fail to capture the complex underlying distributions of multimodal health data and long-term dependencies throughout medical histories. Recent advances in generative artificial intelligence (AI) have provided powerful tools to represent complex distributions and patterns with minimal underlying assumptions, with major impact in fields such as finance and environmental sciences, prompting researchers to apply these methods for disease modeling in health care. OBJECTIVE While AI methods have proven powerful, their application in clinical practice remains limited due to their highly complex nature. The proliferation of AI algorithms also poses a significant challenge for nondevelopers to track and incorporate these advances into clinical research and application. In this paper, we introduce basic concepts in generative AI and discuss current algorithms and how they can be applied to health care for practitioners with little background in computer science. METHODS We surveyed peer-reviewed papers on generative AI models with specific applications to time-series health data. Our search included single- and multimodal generative AI models that operated over structured and unstructured data, physiological waveforms, medical imaging, and multi-omics data. We introduce current generative AI methods, review their applications, and discuss their limitations and future directions in each data modality. RESULTS We followed the PRISMA-ScR (Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews) guidelines and reviewed 155 articles on generative AI applications to time-series health care data across modalities. Furthermore, we offer a systematic framework for clinicians to easily identify suitable AI methods for their data and task at hand. CONCLUSIONS We reviewed and critiqued existing applications of generative AI to time-series health data with the aim of bridging the gap between computational methods and clinical application. We also identified the shortcomings of existing approaches and highlighted recent advances in generative AI that represent promising directions for health care modeling.
Collapse
Affiliation(s)
- Rosemary He
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Varuni Sarwal
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Xinru Qiu
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, United States
| | - Yongwen Zhuang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, United States
| | - Le Zhang
- Institute for Integrative Genome Biology, University of California Riverside, Riverside, CA, United States
| | - Yue Liu
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, United States
| | - Jeffrey Chiang
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
8
|
Heinolainen A, Nguyen B, Silén S, Renkonen R, Koskinen M. Survival and data-driven phenotypes in head and neck cancer. Sci Rep 2025; 15:5985. [PMID: 39966526 PMCID: PMC11836055 DOI: 10.1038/s41598-025-89053-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
Head and neck cancer (HNC) is the seventh most common cancer worldwide, with a 5-year overall survival of 50-60%. Despite known survival factors, unexpected deaths underscore the need to refine pre-treatment survival predictions. This study aimed to identify novel data-driven HNC patient phenotypes, associated features and predict overall survival using deep survival clustering model VaDeSC. We used a retrospective cohort of 1341 HNC patients from Helsinki University Hospital, utilizing their pre-treatment clinical and demographic data from electronic health records. We identified six previously unrecognized HNC phenotypes with distinct survival patterns, highlighting the significance of BMI, sleep apnoea, TNM stage, tumour site, treatment intention, gender, and age-related treatment modalities. VaDeSC demonstrated strong predictive accuracy, clustering performance and generalizability, achieving C-index of 0.895 ± 0.015 on training and 0.782 ± 0.013 on test data. The phenotype purity was 0.102 ± 0.11 on training-validation data and 0.143 ± 0.006 on test data. Our findings demonstrated that clustering pre-treatment clinical survival data offers intuitively interpretable results with accurate individualised predictions. This approach holds significant potential for novel phenotype discovery across various diseases and endpoints.
Collapse
Affiliation(s)
- Anni Heinolainen
- Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Diagnostic Center, Helsinki University Hospital, Helsinki, Finland.
| | - Bruce Nguyen
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Suvi Silén
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Otorhinolaryngology-Head and Neck Surgery, Helsinki University Hospital, Helsinki, Finland
| | - Risto Renkonen
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Miika Koskinen
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
9
|
de Swart WK, Loog M, Krijthe JH. A comparative study of methods for dynamic survival analysis. Front Neurol 2025; 16:1504535. [PMID: 40040908 PMCID: PMC11876041 DOI: 10.3389/fneur.2025.1504535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/14/2025] [Indexed: 03/06/2025] Open
Abstract
Introduction Dynamic survival analysis has become an effective approach for predicting time-to-event outcomes based on longitudinal data in neurology, cognitive health, and other health-related domains. With advancements in machine learning, several new methods have been introduced, often using a two-stage approach: first extracting features from longitudinal trajectories and then using these to predict survival probabilities. Methods This work compares several combinations of longitudinal and survival models, assessing their predictive performance across different training strategies. Using synthetic and real-world cognitive health data from the Alzheimer's Disease Neuroimaging Initiative (ADNI), we explore the strengths and limitations of each model. Results Among the considered survival models, the Random Survival Forest consistently delivered strong results across different datasets, longitudinal models, and training strategies. On the ADNI dataset the best performing method was Random Survival Forest with the last visit benchmark and super landmarking with an average tdAUC of 0.96 and brier score of 0.07. Several other methods, including Cox Proportional Hazards and the Recurrent Neural Network, achieve similar scores. While the tested longitudinal models often struggled to outperform simple benchmarks, neural network models showed some improvement in simulated scenarios with sufficiently informative longitudinal trajectories. Discussion Our findings underscore the importance of aligning model selection and training strategies with the specific characteristics of the data and the target application, providing valuable insights that can inform future developments in dynamic survival analysis.
Collapse
Affiliation(s)
- Wieske K. de Swart
- Institute for Computing and Information Sciences, Radboud University, Nijmegen, Netherlands
| | - Marco Loog
- Institute for Computing and Information Sciences, Radboud University, Nijmegen, Netherlands
| | - Jesse H. Krijthe
- Institute for Computing and Information Sciences, Radboud University, Nijmegen, Netherlands
- Pattern Recognition Laboratory, Delft University of Technology, Delft, Netherlands
| |
Collapse
|
10
|
Shen L, Zhang T, Xu J, Jiang Y, Cao F, Chen Q, Li C, Nuerhashi G, Li W, Wu P, Fan W. Survival path model outperforms conventional static machine learning models in long-term dynamic prognosis prediction for patients with intermediate stage hepatocellular carcinoma. BIOINFORMATICS ADVANCES 2025; 5:vbaf027. [PMID: 40201235 PMCID: PMC11978388 DOI: 10.1093/bioadv/vbaf027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/09/2024] [Accepted: 02/13/2025] [Indexed: 04/10/2025]
Abstract
Motivation Patients with intermediate stage hepatocellular carcinoma (HCC) require repeated disease monitoring, prognosis assessment, and treatment planning. A novel machine learning model called survival path mapping (SP) model was developed, while its performance as compared with conventional machine learning models remains unknown. Between January 2007 and December 2018, the time-series data of 2644 intermediate stage HCC patients from four medical centers in China were reviewed and included. Static machine learning models by Gaussian Naive Bayes (GNB), support vector machine (SVM), and random forest (RF) for the prediction of survivorship were built based on data at initial admission. Longitudinal data divided into different time slices were utilized for the construction of the SP model. The time-dependent c-index was compared between models. Results The training set, internal testing set, and external testing set consisted of 1560, 670, and 414 HCC patients, respectively. The survival path model had superior or non-inferior performance in prognosis prediction compared to GNB and RF models since the 12th month after initial diagnosis in the training set and the external testing set. The survival path model had higher time-dependent c-index over all conventional ML models since the 6th month in the external testing cohort. In conclusion, the survival path model had superior performance in long-term dynamic prognosis prediction compared to conventional static machine learning models for intermediate stage HCC. Availability and implementation The parameters of models are provided in the manuscript.
Collapse
Affiliation(s)
- Lujun Shen
- Department of Minimally Invasive Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Tao Zhang
- Department of Information, Nanfang Hospital, Southern Medical University, Guangzhou 510060, P. R. China
| | - Jian Xu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, P. R. China
| | - Yiquan Jiang
- Department of Minimally Invasive Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Fei Cao
- Department of Minimally Invasive Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Qifeng Chen
- Department of Minimally Invasive Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Chen Li
- Department of Minimally Invasive Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Gulijiayina Nuerhashi
- Department of Minimally Invasive Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Wang Li
- Department of Minimally Invasive Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Peihong Wu
- Department of Minimally Invasive Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Weijun Fan
- Department of Minimally Invasive Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| |
Collapse
|
11
|
Wang Y, Gupta A, Tushar FI, Riley B, Wang A, Tailor TD, Tantum S, Liu JG, Bashir MR, Lo JY, Lafata KJ. Concordance-based Predictive Uncertainty (CPU)-Index: Proof-of-concept with application towards improved specificity of lung cancers on low dose screening CT. Artif Intell Med 2025; 160:103055. [PMID: 39721356 DOI: 10.1016/j.artmed.2024.103055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
In this paper, we introduce a novel concordance-based predictive uncertainty (CPU)-Index, which integrates insights from subgroup analysis and personalized AI time-to-event models. Through its application in refining lung cancer screening (LCS) predictions generated by an individualized AI time-to-event model trained with fused data of low dose CT (LDCT) radiomics with patient demographics, we demonstrate its effectiveness, resulting in improved risk assessment compared to the Lung CT Screening Reporting & Data System (Lung-RADS). Subgroup-based Lung-RADS faces challenges in representing individual variations and relies on a limited set of predefined characteristics, resulting in variable predictions. Conversely, personalized AI time-to-event models are hindered by transparency issues and biases from censored data. By measuring the prediction consistency between subgroup analysis and AI time-to-event models, the CPU-Index framework offers a nuanced evaluation of the bias-variance trade-off and improves the transparency and reliability of predictions. Consistency was estimated by the concordance index of subgroup analysis-based similarity rank and model prediction similarity rank. Subgroup analysis-based similarity loss was defined as the sum-of-the-difference between Lung-RADS and feature-level 0-1 loss. Model prediction similarity loss was defined as squared loss. To test our approach, we identified 3,326 patients who underwent LDCT for LCS from 1/1/2015 to 6/30/2020 with confirmation of lung cancer on pathology within one year. For each LDCT image, the lesion associated with a Lung-RADS score was detected using a pretrained deep learning model from Medical Open Network for AI (MONAI), from which radiomic features were extracted. Radiomics were optimally fused with patient demographics via a positional encoding scheme and used to train a neural multi-task logistic regression time-to-event model that predicts malignancy. Performance was maximized when radiomics features were fused with positionally encoded demographic features. In this configuration, our algorithm raised the AUC from 0.81 ± 0.04 to 0.89 ± 0.02. Compared to standard Lung-RADS, our approach reduced the False-Positive-Rate from 0.41 ± 0.02 to 0.30 ± 0.12 while maintaining the same False-Negative-Rate. Our methodology enhances lung cancer risk assessment by estimating prediction uncertainty and adjusting accordingly. Furthermore, the optimal integration of radiomics and patient demographics improved overall diagnostic performance, indicating their complementary nature.
Collapse
Affiliation(s)
- Yuqi Wang
- Department of Electrical and Computer Engineering, Duke University, Durham, NC, United States of America.
| | - Aarzu Gupta
- Department of Electrical and Computer Engineering, Duke University, Durham, NC, United States of America
| | - Fakrul Islam Tushar
- Department of Electrical and Computer Engineering, Duke University, Durham, NC, United States of America
| | - Breylon Riley
- Medical Physics Graduate Program, Duke University, Durham, NC, United States of America
| | - Avivah Wang
- School of Medicine, Duke University, Durham, NC, United States of America
| | - Tina D Tailor
- Department of Radiology, Duke University, Durham, NC, United States of America
| | - Stacy Tantum
- Department of Electrical and Computer Engineering, Duke University, Durham, NC, United States of America
| | - Jian-Guo Liu
- Departments of Physics and Mathematics, Duke University, Durham, NC, United States of America
| | - Mustafa R Bashir
- Department of Radiology, Duke University, Durham, NC, United States of America; Department of Medicine, Division of Gastroenterology, Duke University, Durham, NC, United States of America
| | - Joseph Y Lo
- Department of Electrical and Computer Engineering, Duke University, Durham, NC, United States of America; Medical Physics Graduate Program, Duke University, Durham, NC, United States of America; Department of Radiology, Duke University, Durham, NC, United States of America; Department of Biomedical Engineering, Duke University, Durham, NC, United States of America
| | - Kyle J Lafata
- Department of Electrical and Computer Engineering, Duke University, Durham, NC, United States of America; Medical Physics Graduate Program, Duke University, Durham, NC, United States of America; Department of Radiology, Duke University, Durham, NC, United States of America; Department of Biomedical Engineering, Duke University, Durham, NC, United States of America; Department of Radiation Oncology, Duke University, Durham, NC, United States of America; Department of Pathology, Duke University, Durham, NC, United States of America.
| |
Collapse
|
12
|
Li F, Sun Z, abdelhameed A, Duan T, Rasmy L, Hu X, He J, Dang Y, Feng J, Li J, Wang Y, Lyu T, Braun N, Pham S, Gharacholou M, Fairweather D, Zhi D, Bian J, Tao C. Contrastive learning with transformer for adverse endpoint prediction in patients on DAPT post-coronary stent implantation. Front Cardiovasc Med 2025; 11:1460354. [PMID: 39872877 PMCID: PMC11769931 DOI: 10.3389/fcvm.2024.1460354] [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: 07/05/2024] [Accepted: 12/19/2024] [Indexed: 01/30/2025] Open
Abstract
Background Effective management of dual antiplatelet therapy (DAPT) following drug-eluting stent (DES) implantation is crucial for preventing adverse events. Traditional prognostic tools, such as rule-based methods or Cox regression, despite their widespread use and ease, tend to yield moderate predictive accuracy within predetermined timeframes. This study introduces a new contrastive learning-based approach to enhance prediction efficacy over multiple time intervals. Methods We utilized retrospective, real-world data from the OneFlorida + Clinical Research Consortium. Our study focused on two primary endpoints: ischemic and bleeding events, with prediction windows of 1, 2, 3, 6, and 12 months post-DES implantation. Our approach first utilized an auto-encoder to compress patient features into a more manageable, condensed representation. Following this, we integrated a Transformer architecture with multi-head attention mechanisms to focus on and amplify the most salient features, optimizing the representation for better predictive accuracy. Then, we applied contrastive learning to enable the model to further refine its predictive capabilities by maximizing intra-class similarities and distinguishing inter-class differences. Meanwhile, the model was holistically optimized using multiple loss functions, to ensure the predicted results closely align with the ground-truth values from various perspectives. We benchmarked model performance against three cutting-edge deep learning-based survival models, i.e., DeepSurv, DeepHit, and SurvTrace. Results The final cohort comprised 19,713 adult patients who underwent DES implantation with more than 1 month of records after coronary stenting. Our approach demonstrated superior predictive performance for both ischemic and bleeding events across prediction windows of 1, 2, 3, 6, and 12 months, with time-dependent concordance (Ctd) index values ranging from 0.88 to 0.80 and 0.82 to 0.77, respectively. It consistently outperformed the baseline models, including DeepSurv, DeepHit, and SurvTrace, with statistically significant improvement in the Ctd-index values for most evaluated scenarios. Conclusion The robust performance of our contrastive learning-based model underscores its potential to enhance DAPT management significantly. By delivering precise predictive insights at multiple time points, our method meets the current need for adaptive, personalized therapeutic strategies in cardiology, thereby offering substantial value in improving patient outcomes.
Collapse
Affiliation(s)
- Fang Li
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL, United States
| | - Zenan Sun
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Ahmed abdelhameed
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL, United States
| | - Tiehang Duan
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL, United States
| | - Laila Rasmy
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Xinyue Hu
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL, United States
| | - Jianping He
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yifang Dang
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jingna Feng
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL, United States
| | - Jianfu Li
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL, United States
| | - Yichen Wang
- Division of Hospital Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Tianchen Lyu
- Health Outcomes and Biomedical Informatics, University of Florida Health, Gainesville, FL, United States
| | - Naomi Braun
- Health Outcomes and Biomedical Informatics, University of Florida Health, Gainesville, FL, United States
| | - Si Pham
- Department of Cardiothoracic Surgery, Mayo Clinic, Jacksonville, FL, United States
| | - Michael Gharacholou
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Degui Zhi
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jiang Bian
- Health Outcomes and Biomedical Informatics, University of Florida Health, Gainesville, FL, United States
| | - Cui Tao
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
13
|
Zeng L, Zhang J, Chen W, Ding Y. tdCoxSNN: Time-dependent Cox survival neural network for continuous-time dynamic prediction. J R Stat Soc Ser C Appl Stat 2025; 74:187-203. [PMID: 39807175 PMCID: PMC11725344 DOI: 10.1093/jrsssc/qlae051] [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: 01/12/2024] [Revised: 06/22/2024] [Accepted: 09/15/2024] [Indexed: 01/16/2025]
Abstract
The aim of dynamic prediction is to provide individualized risk predictions over time, which are updated as new data become available. In pursuit of constructing a dynamic prediction model for a progressive eye disorder, age-related macular degeneration (AMD), we propose a time-dependent Cox survival neural network (tdCoxSNN) to predict its progression using longitudinal fundus images. tdCoxSNN builds upon the time-dependent Cox model by utilizing a neural network to capture the nonlinear effect of time-dependent covariates on the survival outcome. Moreover, by concurrently integrating a convolutional neural network with the survival network, tdCoxSNN can directly take longitudinal images as input. We evaluate and compare our proposed method with joint modelling and landmarking approaches through extensive simulations. We applied the proposed approach to two real datasets. One is a large AMD study, the Age-Related Eye Disease Study, in which more than 50,000 fundus images were captured over a period of 12 years for more than 4,000 participants. Another is a public dataset of the primary biliary cirrhosis disease, where multiple laboratory tests were longitudinally collected to predict the time-to-liver transplant. Our approach demonstrates commendable predictive performance in both simulation studies and the analysis of the two real datasets.
Collapse
Affiliation(s)
- Lang Zeng
- Department of Biostatistics and Health Data Science, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jipeng Zhang
- Department of Biostatistics and Health Data Science, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wei Chen
- Department of Biostatistics and Health Data Science, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ying Ding
- Department of Biostatistics and Health Data Science, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Xu Z, Scharp D, Hobensack M, Ye J, Zou J, Ding S, Shang J, Topaz M. Machine learning-based infection diagnostic and prognostic models in post-acute care settings: a systematic review. J Am Med Inform Assoc 2025; 32:241-252. [PMID: 39530740 PMCID: PMC11648729 DOI: 10.1093/jamia/ocae278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVES This study aims to (1) review machine learning (ML)-based models for early infection diagnostic and prognosis prediction in post-acute care (PAC) settings, (2) identify key risk predictors influencing infection-related outcomes, and (3) examine the quality and limitations of these models. MATERIALS AND METHODS PubMed, Web of Science, Scopus, IEEE Xplore, CINAHL, and ACM digital library were searched in February 2024. Eligible studies leveraged PAC data to develop and evaluate ML models for infection-related risks. Data extraction followed the CHARMS checklist. Quality appraisal followed the PROBAST tool. Data synthesis was guided by the socio-ecological conceptual framework. RESULTS Thirteen studies were included, mainly focusing on respiratory infections and nursing homes. Most used regression models with structured electronic health record data. Since 2020, there has been a shift toward advanced ML algorithms and multimodal data, biosensors, and clinical notes being significant sources of unstructured data. Despite these advances, there is insufficient evidence to support performance improvements over traditional models. Individual-level risk predictors, like impaired cognition, declined function, and tachycardia, were commonly used, while contextual-level predictors were barely utilized, consequently limiting model fairness. Major sources of bias included lack of external validation, inadequate model calibration, and insufficient consideration of data complexity. DISCUSSION AND CONCLUSION Despite the growth of advanced modeling approaches in infection-related models in PAC settings, evidence supporting their superiority remains limited. Future research should leverage a socio-ecological lens for predictor selection and model construction, exploring optimal data modalities and ML model usage in PAC, while ensuring rigorous methodologies and fairness considerations.
Collapse
Affiliation(s)
- Zidu Xu
- School of Nursing, Columbia University, New York, NY 10032,
United States
| | - Danielle Scharp
- School of Nursing, Columbia University, New York, NY 10032,
United States
| | - Mollie Hobensack
- Icahn School of Medicine at Mount Sinai, New York, NY 10029,
United States
| | - Jiancheng Ye
- Weill Cornell Medicine, Cornell University, New York, NY
10065, United States
| | - Jungang Zou
- Department of Biostatistics, Mailman School of Public Health, Columbia
University, New York, NY 10032, United
States
| | - Sirui Ding
- Bakar Computational Health Sciences Institute, University of
California, San Francisco, CA 94158, United
States
| | - Jingjing Shang
- School of Nursing, Columbia University, New York, NY 10032,
United States
| | - Maxim Topaz
- School of Nursing, Columbia University, New York, NY 10032,
United States
- Center for Home Care Policy & Research, VNS Health, New
York, NY 10001, United States
- Data Science Institute, Columbia University, New York, NY
10027, United States
| |
Collapse
|
15
|
Buttar AM, Shaheen Z, Gumaei AH, Mosleh MAA, Gupta I, Alzanin SM, Akbar MA. Enhanced neurological anomaly detection in MRI images using deep convolutional neural networks. Front Med (Lausanne) 2024; 11:1504545. [PMID: 39802885 PMCID: PMC11717658 DOI: 10.3389/fmed.2024.1504545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Introduction Neurodegenerative diseases, including Parkinson's, Alzheimer's, and epilepsy, pose significant diagnostic and treatment challenges due to their complexity and the gradual degeneration of central nervous system structures. This study introduces a deep learning framework designed to automate neuro-diagnostics, addressing the limitations of current manual interpretation methods, which are often time-consuming and prone to variability. Methods We propose a specialized deep convolutional neural network (DCNN) framework aimed at detecting and classifying neurological anomalies in MRI data. Our approach incorporates key preprocessing techniques, such as reducing noise and normalizing image intensity in MRI scans, alongside an optimized model architecture. The model employs Rectified Linear Unit (ReLU) activation functions, the Adam optimizer, and a random search strategy to fine-tune hyper-parameters like learning rate, batch size, and the number of neurons in fully connected layers. To ensure reliability and broad applicability, cross-fold validation was used. Results and discussion Our DCNN achieved a remarkable classification accuracy of 98.44%, surpassing well-known models such as ResNet-50 and AlexNet when evaluated on a comprehensive MRI dataset. Moreover, performance metrics such as precision, recall, and F1-score were calculated separately, confirming the robustness and efficiency of our model across various evaluation criteria. Statistical analyses, including ANOVA and t-tests, further validated the significance of the performance improvements observed with our proposed method. This model represents an important step toward creating a fully automated system for diagnosing and planning treatment for neurological diseases. The high accuracy of our framework highlights its potential to improve diagnostic workflows by enabling precise detection, tracking disease progression, and supporting personalized treatment strategies. While the results are promising, further research is necessary to assess how the model performs across different clinical scenarios. Future studies could focus on integrating additional data types, such as longitudinal imaging and multimodal techniques, to further enhance diagnostic accuracy and clinical utility. These findings mark a significant advancement in applying deep learning to neuro-diagnostics, with promising implications for improving patient outcomes and clinical practices.
Collapse
Affiliation(s)
- Ahmed Mateen Buttar
- Department of Computer Science, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Zubair Shaheen
- Department of Computer Science, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Abdu H. Gumaei
- Department of Computer Science, College of Computer Engineering and Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mogeeb A. A. Mosleh
- Faculty of Engineering and Information Technology, Taiz University, Taiz, Yemen
- Faculty of Engineering and Computing, University of Science and Technology, Aden, Yemen
| | - Indrajeet Gupta
- School of Computer Science & AI SR University, Warangal, Telangana, India
| | - Samah M. Alzanin
- Department of Computer Science, College of Computer Engineering and Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | |
Collapse
|
16
|
Sun J, Deng L, Li Q, Zhou J, Zhang Y. Dynamic relations between longitudinal morphological, behavioral, and emotional indicators and cognitive impairment: evidence from the Chinese Longitudinal Healthy Longevity Survey. BMC Public Health 2024; 24:3516. [PMID: 39696204 DOI: 10.1186/s12889-024-21072-w] [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: 09/04/2023] [Accepted: 12/11/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND We aimed to assess the effects of body mass index (BMI), activities of daily living (ADL), and subjective well-being (SWB) on cognitive impairment and propose dynamic risk prediction models for aging cognitive decline. METHODS We leveraged the Chinese Longitudinal Healthy Longevity Survey from 1998 to 2018. Cognitive status was measured using the Chinese Mini-Mental State Examination. We employed repeated measures correlation to assess associations, linear mixed-effect models to characterize the longitudinal changes, and Cox proportional hazard regression to model survival time. Dynamic predictive models were established based on the Bayesian joint model and deep learning approach named dynamic-DeepHit. Marginal structural Cox models were adopted to control for time-varying confounding factors and assess effect sizes. RESULTS ADL, SWB, and BMI showed protective effects on cognitive impairment after controlling observed confounding factors, with respective direct hazard ratios of 0.756 (0.741, 0.771), 0.912 (0.902, 0.921), and 0.919 (0.909, 0.929). Dynamic risk predictive models manifested high accuracy (best AUC = 0.89). ADL was endowed with the best predictive capability, although the combination of BMI, ADL, and SWB showed the most remarkable performance. CONCLUSIONS BMI, ADL, and SWB are protective factors for cognitive impairment. A dynamic prediction model using these indicators can efficiently identify vulnerable individuals with high accuracy.
Collapse
Affiliation(s)
- Jianle Sun
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
- Department of Philosophy, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Luojia Deng
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qianwen Li
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jie Zhou
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yue Zhang
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
17
|
Periáñez Á, Fernández Del Río A, Nazarov I, Jané E, Hassan M, Rastogi A, Tang D. The Digital Transformation in Health: How AI Can Improve the Performance of Health Systems. Health Syst Reform 2024; 10:2387138. [PMID: 39437247 DOI: 10.1080/23288604.2024.2387138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/27/2024] [Accepted: 07/29/2024] [Indexed: 10/25/2024] Open
Abstract
Mobile health has the potential to revolutionize health care delivery and patient engagement. In this work, we discuss how integrating Artificial Intelligence into digital health applications focused on supply chain operation, patient management, and capacity building, among other use cases, can improve the health system and public health performance. We present the Causal Foundry Artificial Intelligence and Reinforcement Learning platform, which allows the delivery of adaptive interventions whose impact can be optimized through experimentation and real-time monitoring. The system can integrate multiple data sources and digital health applications. The flexibility of this platform to connect to various mobile health applications and digital devices, and to send personalized recommendations based on past data and predictions, can significantly improve the impact of digital tools on health system outcomes. The potential for resource-poor settings, where the impact of this approach on health outcomes could be decisive, is discussed. This framework is similarly applicable to improving efficiency in health systems where scarcity is not an issue.
Collapse
|
18
|
Medina-Olivares V, Lessmann S, Klein N. The Deep Promotion Time Cure Model. IEEE TRANSACTIONS ON NEURAL NETWORKS AND LEARNING SYSTEMS 2024; 35:18848-18858. [PMID: 38771686 DOI: 10.1109/tnnls.2024.3398559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
We propose a novel method for predicting time-to-event data in the presence of cure fractions based on flexible survival models integrated into a deep neural network (DNN) framework. Our approach allows for nonlinear relationships and high-dimensional interactions between covariates and survival and is suitable for large-scale applications. To ensure the identifiability of the overall predictor formed of an additive decomposition of interpretable linear and nonlinear effects and potential higher-dimensional interactions captured through a DNN, we employ an orthogonalization layer. We demonstrate the usefulness and computational efficiency of our method via simulations and apply it to a large portfolio of U.S. mortgage loans. Here, we find not only a better predictive performance of our framework but also a more realistic picture of covariate effects.
Collapse
|
19
|
Mesinovic M, Watkinson P, Zhu T. DySurv: dynamic deep learning model for survival analysis with conditional variational inference. J Am Med Inform Assoc 2024:ocae271. [PMID: 39569428 DOI: 10.1093/jamia/ocae271] [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: 02/14/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 11/22/2024] Open
Abstract
OBJECTIVE Machine learning applications for longitudinal electronic health records often forecast the risk of events at fixed time points, whereas survival analysis achieves dynamic risk prediction by estimating time-to-event distributions. Here, we propose a novel conditional variational autoencoder-based method, DySurv, which uses a combination of static and longitudinal measurements from electronic health records to estimate the individual risk of death dynamically. MATERIALS AND METHODS DySurv directly estimates the cumulative risk incidence function without making any parametric assumptions on the underlying stochastic process of the time-to-event. We evaluate DySurv on 6 time-to-event benchmark datasets in healthcare, as well as 2 real-world intensive care unit (ICU) electronic health records (EHR) datasets extracted from the eICU Collaborative Research (eICU) and the Medical Information Mart for Intensive Care database (MIMIC-IV). RESULTS DySurv outperforms other existing statistical and deep learning approaches to time-to-event analysis across concordance and other metrics. It achieves time-dependent concordance of over 60% in the eICU case. It is also over 12% more accurate and 22% more sensitive than in-use ICU scores like Acute Physiology and Chronic Health Evaluation (APACHE) and Sequential Organ Failure Assessment (SOFA) scores. The predictive capacity of DySurv is consistent and the survival estimates remain disentangled across different datasets. DISCUSSION Our interdisciplinary framework successfully incorporates deep learning, survival analysis, and intensive care to create a novel method for time-to-event prediction from longitudinal health records. We test our method on several held-out test sets from a variety of healthcare datasets and compare it to existing in-use clinical risk scoring benchmarks. CONCLUSION While our method leverages non-parametric extensions to deep learning-guided estimations of the survival distribution, further deep learning paradigms could be explored.
Collapse
Affiliation(s)
- Munib Mesinovic
- Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, United Kingdom
| | - Peter Watkinson
- Critical Care Research Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 7JX, United Kingdom
| | - Tingting Zhu
- Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, United Kingdom
| |
Collapse
|
20
|
Li LY, Isaksen AA, Lebiecka-Johansen B, Funck K, Thambawita V, Byberg S, Andersen TH, Norgaard O, Hulman A. Prediction of cardiovascular markers and diseases using retinal fundus images and deep learning: a systematic scoping review. EUROPEAN HEART JOURNAL. DIGITAL HEALTH 2024; 5:660-669. [PMID: 39563905 PMCID: PMC11570365 DOI: 10.1093/ehjdh/ztae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/28/2024] [Accepted: 09/05/2024] [Indexed: 11/21/2024]
Abstract
Rapid development in deep learning for image analysis inspired studies to focus on predicting cardiovascular risk using retinal fundus images. This scoping review aimed to identify and describe studies using retinal fundus images and deep learning to predict cardiovascular risk markers and diseases. We searched MEDLINE and Embase on 17 November 2023. Abstracts and relevant full-text articles were independently screened by two reviewers. We included studies that used deep learning for the analysis of retinal fundus images to predict cardiovascular risk markers or cardiovascular diseases (CVDs) and excluded studies only using predefined characteristics of retinal fundus images. Study characteristics were presented using descriptive statistics. We included 24 articles published between 2018 and 2023. Among these, 23 (96%) were cross-sectional studies and eight (33%) were follow-up studies with clinical CVD outcomes. Seven studies included a combination of both designs. Most studies (96%) used convolutional neural networks to process images. We found nine (38%) studies that incorporated clinical risk factors in the prediction and four (17%) that compared the results to commonly used clinical risk scores in a prospective setting. Three of these reported improved discriminative performance. External validation of models was rare (21%). There is increasing interest in using retinal fundus images in cardiovascular risk assessment with some studies demonstrating some improvements in prediction. However, more prospective studies, comparisons of results to clinical risk scores, and models augmented with traditional risk factors can strengthen further research in the field.
Collapse
Affiliation(s)
- Livie Yumeng Li
- Department of Public Health, Aarhus University, Bartholins Allé 2, 8000 Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
| | - Anders Aasted Isaksen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
| | - Benjamin Lebiecka-Johansen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
| | - Kristian Funck
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
| | - Vajira Thambawita
- Department of Holistic Systems, SimulaMet, Stensberggata 27, 0170 Oslo, Norway
| | - Stine Byberg
- Clinical Epidemiological Research, Copenhagen University Hospital — Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark
| | - Tue Helms Andersen
- Department of Education, Danish Diabetes Knowledge Center, Copenhagen University Hospital — Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark
| | - Ole Norgaard
- Department of Education, Danish Diabetes Knowledge Center, Copenhagen University Hospital — Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark
| | - Adam Hulman
- Department of Public Health, Aarhus University, Bartholins Allé 2, 8000 Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
| |
Collapse
|
21
|
Zheng Q, Shen Q, Shu Z, Chang K, Zhong K, Yan Y, Ke J, Huang J, Su R, Xia J, Zhou X. Deep representation learning from electronic medical records identifies distinct symptom based subtypes and progression patterns for COVID-19 prognosis. Int J Med Inform 2024; 191:105555. [PMID: 39089210 DOI: 10.1016/j.ijmedinf.2024.105555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/17/2024] [Accepted: 07/14/2024] [Indexed: 08/03/2024]
Abstract
OBJECTIVE Symptoms are significant kind of phenotypes for managing and controlling of the burst of acute infectious diseases, such as COVID-19. Although patterns of symptom clusters and time series have been considered the high potential prediction factors for the prognosis of patients, the elaborated subtypes and their progression patterns based on symptom phenotypes related to the prognosis of COVID-19 patients still need be detected. This study aims to investigate patient subtypes and their progression patterns with distinct features of outcome and prognosis. METHODS This study included a total of 14,139 longitudinal electronic medical records (EMRs) obtained from four hospitals in Hubei Province, China, involving 2,683 individuals in the early stage of COVID-19 pandemic. A deep representation learning model was developed to help acquire the symptom profiles of patients. K-means clustering algorithm is used to divide them into distinct subtypes. Subsequently, symptom progression patterns were identified by considering the subtypes associated with patients upon admission and discharge. Furthermore, we used Fisher's test to identify significant clinical entities for each subtype. RESULTS Three distinct patient subtypes exhibiting specific symptoms and prognosis have been identified. Particularly, Subtype 0 includes 44.2% of the whole and is characterized by poor appetite, fatigue and sleep disorders; Subtype 1 includes 25.6% cases and is characterized by confusion, cough with bloody sputum, encopresis and urinary incontinence; Subtype 2 includes 30.2% cases and is characterized by dry cough and rhinorrhea. These three subtypes demonstrate significant disparities in prognosis, with the mortality rates of 4.72%, 8.59%, and 0.25% respectively. Furthermore, symptom cluster progression patterns showed that patients with Subtype 0 who manifest dark yellow urine, chest pain, etc. in the admission stage exhibit an elevated risk of transforming into the more severe subtypes with poor outcome, whereas those presenting with nausea and vomiting tend to incline towards entering the milder subtype. CONCLUSION This study has proposed a clinical meaningful approach by utilizing the deep representation learning and real-world EMR data containing symptom phenotypes to identify the COVID-19 subtypes and their progression patterns. The results would be potentially useful to help improve the precise stratification and management of acute infectious diseases.
Collapse
Affiliation(s)
- Qiguang Zheng
- School of Computer and Information Technology, Beijing Jiaotong University, China
| | - Qifan Shen
- School of Computer and Information Technology, Beijing Jiaotong University, China
| | - Zixin Shu
- School of Computer and Information Technology, Beijing Jiaotong University, China
| | - Kai Chang
- School of Computer and Information Technology, Beijing Jiaotong University, China
| | - Kunyu Zhong
- School of Computer and Information Technology, Beijing Jiaotong University, China
| | - Yuhang Yan
- School of Computer and Information Technology, Beijing Jiaotong University, China
| | - Jia Ke
- Hubei Provincial Hospital of Traditional Chinese Medicine, China
| | - Jingjing Huang
- Hubei Provincial Hospital of Traditional Chinese Medicine, China
| | - Rui Su
- Beijing Hospital of Traditional Chinese Medicine, China
| | - Jianan Xia
- School of Computer and Information Technology, Beijing Jiaotong University, China.
| | - Xuezhong Zhou
- School of Computer and Information Technology, Beijing Jiaotong University, China.
| |
Collapse
|
22
|
Shen L, Jiang Y, Zhang T, Cao F, Ke L, Li C, Nuerhashi G, Li W, Wu P, Li C, Zeng Q, Fan W. Machine Learning for Dynamic Prognostication of Patients With Hepatocellular Carcinoma Using Time-Series Data: Survival Path Versus Dynamic-DeepHit HCC Model. Cancer Inform 2024; 23:11769351241289719. [PMID: 39421722 PMCID: PMC11483769 DOI: 10.1177/11769351241289719] [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: 05/28/2024] [Accepted: 09/11/2024] [Indexed: 10/19/2024] Open
Abstract
Objectives Patients with intermediate or advanced hepatocellular carcinoma (HCC) require repeated disease monitoring, prognosis assessment and treatment planning. In 2018, a novel machine learning methodology "survival path" (SP) was developed to facilitate dynamic prognosis prediction and treatment planning. One year after, a deep learning approach called Dynamic Deephit was developed. The performance of the two state-of-art models in dynamic prognostication have not been compared. Methods We trained and tested the SP and Dynamic DeepHit models in a large cohort of 2511 HCC patients using time-series data. The time-series data were converted into data of time slices, with an interval of three months. The time-dependent c-index for OS at given prediction time (t = 1, 6, 12, 18 months) and evaluation time (∆t = 3, 6, 9, 12, 18, 24, 36, 48 months) were compared. Results The comparison between SP model and Dynamic DeepHit-HCC model showed the latter had significant better performance at the time of initial admission. The time-dependent c-index of Dynamic DeepHit-HCC model gradually decreased with the extension of time (from 0.756 to 0.639 in the training set; from 0.787 to 0.661 in internal testing set; from 0.725 to 0.668 in multicenter testing set); while the time-dependent c-index of SP model displayed an increased trend (from 0.665 to 0.748 in the training set; from 0.608 to 0.743 in internal testing set; from 0.643 to 0.720 in multicenter testing set). When the prediction time comes to 6 months or later since initial treatment, the survival path model outperformed the dynamic DeepHit model at late evaluation times (∆t > 12 months). Conclusions This research highlighted the unique strengths of both models. The SP model had advantage in long term prediction while the Dynamic DeepHit-HCC model had advantages in prediction at near time points. Fine selection of models is needed in dealing with different scenarios.
Collapse
Affiliation(s)
- Lujun Shen
- Department of Minimally invasive therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yiquan Jiang
- Department of Minimally invasive therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Tao Zhang
- Department of Information, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Fei Cao
- Department of Minimally invasive therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Liangru Ke
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Chen Li
- Department of Minimally invasive therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Gulijiayina Nuerhashi
- Department of Minimally invasive therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Wang Li
- Department of Minimally invasive therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Peihong Wu
- Department of Minimally invasive therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Chaofeng Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Information Center, Sun Yat-sen University Cancer Center, Guangdong, China
| | - Qi Zeng
- Cancer center, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Weijun Fan
- Department of Minimally invasive therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| |
Collapse
|
23
|
Zhang T, Huang M, Chen L, Xia Y, Min W, Jiang S. Machine learning and statistical models to predict all-cause mortality in type 2 diabetes: Results from the UK Biobank study. Diabetes Metab Syndr 2024; 18:103135. [PMID: 39413583 DOI: 10.1016/j.dsx.2024.103135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024]
Abstract
AIMS This study aims to compare the performance of contemporary machine learning models with statistical models in predicting all-cause mortality in patients with type 2 diabetes mellitus and to develop a user-friendly mortality risk prediction tool. METHODS A prospective cohort study was conducted including 22,579 people with diabetes from the UK Biobank. Models evaluated include Cox proportional hazards, random survival forests (RSF), gradient boosting (GB) survival, DeepSurv, and DeepHit. RESULTS Over a median follow-up period of 9 years, 2,665 patients died. Machine learning models outperformed the Cox model in the validation dataset, with C-index values of 0.72-0.73 vs. 0.71 for Cox (p < 0.01). Deep learning models, particularly DeepHit, demonstrated superior calibration and achieved lower Brier scores (0.09 vs. 0.10 for Cox, p < 0.05). An online prediction tool based on the DeepHit was developed for patient care: http://123.57.42.89:6006/. CONCLUSIONS Machine learning models performed better than statistical models, highlighting the potential of machine learning techniques for predicting all-cause mortality risk and facilitating personalized healthcare management for individuals with diabetes.
Collapse
Affiliation(s)
- Tingjing Zhang
- School of Public Health, Wannan Medical College, Wuhu, China; Institutes of Brain Science, Wannan Medical College, Wuhu, China
| | - Mingyu Huang
- The Key Laboratory of Intelligent Information Processing, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Liangkai Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Xia
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Weiqing Min
- The Key Laboratory of Intelligent Information Processing, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Shuqiang Jiang
- The Key Laboratory of Intelligent Information Processing, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
24
|
Holste G, Lin M, Zhou R, Wang F, Liu L, Yan Q, Van Tassel SH, Kovacs K, Chew EY, Lu Z, Wang Z, Peng Y. Harnessing the power of longitudinal medical imaging for eye disease prognosis using Transformer-based sequence modeling. NPJ Digit Med 2024; 7:216. [PMID: 39152209 PMCID: PMC11329720 DOI: 10.1038/s41746-024-01207-4] [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: 02/16/2024] [Accepted: 07/29/2024] [Indexed: 08/19/2024] Open
Abstract
Deep learning has enabled breakthroughs in automated diagnosis from medical imaging, with many successful applications in ophthalmology. However, standard medical image classification approaches only assess disease presence at the time of acquisition, neglecting the common clinical setting of longitudinal imaging. For slow, progressive eye diseases like age-related macular degeneration (AMD) and primary open-angle glaucoma (POAG), patients undergo repeated imaging over time to track disease progression and forecasting the future risk of developing a disease is critical to properly plan treatment. Our proposed Longitudinal Transformer for Survival Analysis (LTSA) enables dynamic disease prognosis from longitudinal medical imaging, modeling the time to disease from sequences of fundus photography images captured over long, irregular time periods. Using longitudinal imaging data from the Age-Related Eye Disease Study (AREDS) and Ocular Hypertension Treatment Study (OHTS), LTSA significantly outperformed a single-image baseline in 19/20 head-to-head comparisons on late AMD prognosis and 18/20 comparisons on POAG prognosis. A temporal attention analysis also suggested that, while the most recent image is typically the most influential, prior imaging still provides additional prognostic value.
Collapse
Affiliation(s)
- Gregory Holste
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Mingquan Lin
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Ruiwen Zhou
- Center for Biostatistics and Data Science, Washington University School of Medicine, St. Louis, MO, USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Lei Liu
- Center for Biostatistics and Data Science, Washington University School of Medicine, St. Louis, MO, USA
| | - Qi Yan
- Department of Obstetrics & Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sarah H Van Tassel
- Israel Englander Department of Ophthalmology, Weill Cornell Medicine, New York, NY, USA
| | - Kyle Kovacs
- Israel Englander Department of Ophthalmology, Weill Cornell Medicine, New York, NY, USA
| | - Emily Y Chew
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Zhiyong Lu
- National Center for Biotechnology Information (NCBI), National Library of Medicine (NLM), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Zhangyang Wang
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX, USA.
| | - Yifan Peng
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
25
|
Butner JD, Dogra P, Chung C, Koay EJ, Welsh JW, Hong DS, Cristini V, Wang Z. Hybridizing mechanistic modeling and deep learning for personalized survival prediction after immune checkpoint inhibitor immunotherapy. NPJ Syst Biol Appl 2024; 10:88. [PMID: 39143136 PMCID: PMC11324794 DOI: 10.1038/s41540-024-00415-8] [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: 03/22/2024] [Accepted: 07/29/2024] [Indexed: 08/16/2024] Open
Abstract
We present a study where predictive mechanistic modeling is combined with deep learning methods to predict individual patient survival probabilities under immune checkpoint inhibitor (ICI) immunotherapy. This hybrid approach enables prediction based on both measures that are calculable from mechanistic models of key mechanisms underlying ICI therapy that may not be directly measurable in the clinic and easily measurable quantities or patient characteristics that are not always readily incorporated into predictive mechanistic models. A deep learning time-to-event predictive model trained on a hybrid mechanistic + clinical data set from 93 patients achieved higher per-patient predictive accuracy based on event-time concordance, Brier score, and negative binomial log-likelihood-based criteria than when trained on only mechanistic model-derived values or only clinical data. Feature importance analysis revealed that both clinical and model-derived parameters play prominent roles in increasing prediction accuracy, further supporting the advantage of our hybrid approach.
Collapse
Affiliation(s)
- Joseph D Butner
- Department of Radiation Oncology, 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.
- The Cameron School of Business, University of St. Thomas, Houston, TX, USA.
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Caroline Chung
- Department of Radiation Oncology, 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
| | - Eugene J Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Medical Education, Texas A&M University School of Medicine, Bryan, TX, USA.
| |
Collapse
|
26
|
Hussain Z, De Brouwer E, Boiarsky R, Setty S, Gupta N, Liu G, Li C, Srimani J, Zhang J, Labotka R, Sontag D. Joint AI-driven event prediction and longitudinal modeling in newly diagnosed and relapsed multiple myeloma. NPJ Digit Med 2024; 7:200. [PMID: 39075240 PMCID: PMC11286964 DOI: 10.1038/s41746-024-01189-3] [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: 08/23/2023] [Accepted: 07/15/2024] [Indexed: 07/31/2024] Open
Abstract
Multiple myeloma management requires a balance between maximizing survival, minimizing adverse events to therapy, and monitoring disease progression. While previous work has proposed data-driven models for individual tasks, these approaches fail to provide a holistic view of a patient's disease state, limiting their utility to assist physician decision-making. To address this limitation, we developed a transformer-based machine learning model that jointly (1) predicts progression-free survival (PFS), overall survival (OS), and adverse events (AE), (2) forecasts key disease biomarkers, and (3) assesses the effect of different treatment strategies, e.g., ixazomib, lenalidomide, dexamethasone (IRd) vs lenalidomide, dexamethasone (Rd). Using TOURMALINE trial data, we trained and internally validated our model on newly diagnosed myeloma patients (N = 703) and externally validated it on relapsed and refractory myeloma patients (N = 720). Our model achieved superior performance to a risk model based on the multiple myeloma international staging system (ISS) (p < 0.001, Bonferroni corrected) and comparable performance to survival models trained separately on each task, but unable to forecast biomarkers. Our approach outperformed state-of-the-art deep learning models, tailored towards forecasting, on predicting key disease biomarkers (p < 0.001, Bonferroni corrected). Finally, leveraging our model's capacity to estimate individual-level treatment effects, we found that patients with IgA kappa myeloma appear to benefit the most from IRd. Our study suggests that a holistic assessment of a patient's myeloma course is possible, potentially serving as the foundation for a personalized decision support system.
Collapse
Affiliation(s)
- Zeshan Hussain
- CSAIL, MIT, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | - Cong Li
- Takeda LLC, Cambridge, MA, USA
| | | | | | | | | |
Collapse
|
27
|
Lee D, Kim S, Lee S, Kim HJ, Kim JH, Lim MC, Cho H. Deep Learning-Based Dynamic Risk Prediction of Venous Thromboembolism for Patients With Ovarian Cancer in Real-World Settings From Electronic Health Records. JCO Clin Cancer Inform 2024; 8:e2300192. [PMID: 38996199 DOI: 10.1200/cci.23.00192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/19/2024] [Accepted: 04/16/2024] [Indexed: 07/14/2024] Open
Abstract
PURPOSE Patients with epithelial ovarian cancer (EOC) have an elevated risk for venous thromboembolism (VTE). To assess the risk of VTE, models were developed by statistical or machine learning algorithms. However, few models have accommodated deep learning (DL) algorithms in realistic clinical settings. We aimed to develop a predictive DL model, exploiting rich information from electronic health records (EHRs), including dynamic clinical features and the presence of competing risks. METHODS We extracted EHRs of 1,268 patients diagnosed with EOC from January 2007 through December 2017 at the National Cancer Center, Korea. DL survival networks using fully connected layers, temporal attention, and recurrent neural networks were adopted and compared with multi-perceptron-based classification models. Prediction accuracy was independently validated in the data set of 423 patients newly diagnosed with EOC from January 2018 to December 2019. Personalized risk plots displaying the individual interval risk were developed. RESULTS DL-based survival networks achieved a superior area under the receiver operating characteristic curve (AUROC) between 0.95 and 0.98 while the AUROC of classification models was between 0.85 and 0.90. As clinical information benefits the prediction accuracy, the proposed dynamic survival network outperformed other survival networks for the test and validation data set with the highest time-dependent concordance index (0.974, 0.975) and lowest Brier score (0.051, 0.049) at 6 months after a cancer diagnosis. Our visualization showed that the interval risk fluctuating along with the changes in longitudinal clinical features. CONCLUSION Adaption of dynamic patient clinical features and accounting for competing risks from EHRs into the DL algorithms demonstrated VTE risk prediction with high accuracy. Our results show that this novel dynamic survival network can provide personalized risk prediction with the potential to assist risk-based clinical intervention to prevent VTE among patients with EOC.
Collapse
Affiliation(s)
- Dahhay Lee
- Department of Cancer AI and Digital Health, National Cancer Center Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
- School of Mathematics and Computing (Computational Science and Engineering), Yonsei University, Seoul, Republic of Korea
| | - Seongyoon Kim
- School of Mathematics and Computing (Computational Science and Engineering), Yonsei University, Seoul, Republic of Korea
| | - Sanghee Lee
- Department of Cancer AI and Digital Health, National Cancer Center Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
- Health Insurance Research Institute, National Health Insurance Service, Wonju, Republic of Korea
| | - Hak Jin Kim
- Department of Cardiology, Gumdan Top General Hospital, Incheon, Republic of Korea
- Branch of Cardiology, Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea
| | - Ji Hyun Kim
- Center for Gynecologic Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Myong Cheol Lim
- Center for Gynecologic Cancer, National Cancer Center, Goyang, Republic of Korea
- Division of Tumor Immunology, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
- Center for Clinical Trials, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Hyunsoon Cho
- Department of Cancer AI and Digital Health, National Cancer Center Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
- Integrated Biostatistics Research Branch, National Cancer Center, Goyang, Republic of Korea
| |
Collapse
|
28
|
Yang J, Hu Z, Zhang L, Peng B. Predicting Drugs Suspected of Causing Adverse Drug Reactions Using Graph Features and Attention Mechanisms. Pharmaceuticals (Basel) 2024; 17:822. [PMID: 39065673 PMCID: PMC11279999 DOI: 10.3390/ph17070822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Adverse drug reactions (ADRs) refer to an unintended harmful reaction that occurs after the administration of a medication for therapeutic purposes, which is unrelated to the intended pharmacological action of the drug. In the United States, ADRs account for 6% of all hospital admissions annually. The cost of ADR-related illnesses in 2016 was estimated at USD 528.4 billion. Increasing the awareness of ADRs is an effective measure to prevent them. Assessing suspected drugs in adverse events helps to enhance the awareness of ADRs. METHODS In this study, a suspect drug assisted judgment model (SDAJM) is designed to identify suspected drugs in adverse events. This framework utilizes the graph isomorphism network (GIN) and an attention mechanism to extract features based on patients' demographic information, drug information, and ADR information. RESULTS By comparing it with other models, the results of various tests show that this model performs well in predicting the suspected drugs in adverse reaction events. ADR signal detection was conducted on a group of cardiovascular system drugs, and case analyses were performed on two classic drugs, Mexiletine and Captopril, as well as on two classic antithyroid drugs. The results indicate that the model can accomplish the task of predicting drug ADRs. Validation using benchmark datasets from ten drug discovery domains shows that the model is applicable to classification tasks on the Tox21 and SIDER datasets. CONCLUSIONS This study applies deep learning methods to construct the SDAJM model for three purposes: (1) identifying drugs suspected to cause adverse drug events (ADEs), (2) predicting the ADRs of drugs, and (3) other drug discovery tasks. The results indicate that this method can offer new directions for research in the field of ADRs.
Collapse
Affiliation(s)
| | | | | | - Bin Peng
- College of Public Health, Chongqing Medical University, Chongqing 401331, China; (J.Y.); (Z.H.); (L.Z.)
| |
Collapse
|
29
|
Alter DA, Austin PC, Rosenfeld A. The Dynamic Nature of the Socioeconomic Determinants of Cardiovascular Health: A Narrative Review. Can J Cardiol 2024; 40:989-999. [PMID: 38309464 DOI: 10.1016/j.cjca.2024.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/08/2024] [Accepted: 01/14/2024] [Indexed: 02/05/2024] Open
Abstract
Despite decades of social epidemiologic research, health inequities remain pervasive and ubiquitous in Canada and elsewhere. One reason may be our use of socioeconomic measurement, which has often relied on single point-in-time exposures. To explore the extent to which researchers have incorporated dynamic socioeconomic measurement into cardiovascular health outcome evaluations, we performed a narrative review. We estimated the prevalence of socioeconomic longitudinal cardiovascular research studies that identified socioeconomic exposures at 2 or more points in time between the years of 2019 and 2023. We defined cardiovascular outcome studies as those that examined coronary artery disease, myocardial infarction, acute coronary syndrome, stroke, heart failure, cardiac arrhythmias, cardiac death, cardiometabolic factors, transient ischemic attacks, peripheral artery disease, or hypertension. Socioeconomic exposures included individual income, neighbourhood income, intergenerational social mobility, education, occupation, insurance status, and economic security. Seven percent of socioeconomic cardiovascular outcome studies have measured socioeconomic status at 2 or more points in time throughout the follow-up period, hypothesized mechanisms by which dynamic socioeconomic measures affected outcome focused on social mobility, accumulation, and critical period theories. Insights, implications, and future directions are discussed, in which we highlight ways in which postal code data can be better used methodologically as a dynamic socioeconomic measure. Future research must incorporate dynamic socioeconomic measurement to better inform root causes, interventions, and health-system designs if health equity is to be improved.
Collapse
Affiliation(s)
- David A Alter
- ICES, Sunnybrook Health Sciences, Toronto, Ontario, Canada; Toronto Rehabilitation Institute-University Health Network, Toronto, Ontario, Canada; Institute for Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada.
| | - Peter C Austin
- ICES, Sunnybrook Health Sciences, Toronto, Ontario, Canada; Institute for Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Aaron Rosenfeld
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Wu Q, Dai J. Enhanced osteoporotic fracture prediction in postmenopausal women using Bayesian optimization of machine learning models with genetic risk score. J Bone Miner Res 2024; 39:462-472. [PMID: 38477741 PMCID: PMC11262147 DOI: 10.1093/jbmr/zjae025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 03/14/2024]
Abstract
This study aimed to enhance the fracture risk prediction accuracy in major osteoporotic fractures (MOFs) and hip fractures (HFs) by integrating genetic profiles, machine learning (ML) techniques, and Bayesian optimization. The genetic risk score (GRS), derived from 1,103 risk single nucleotide polymorphisms (SNPs) from genome-wide association studies (GWAS), was formulated for 25,772 postmenopausal women from the Women's Health Initiative dataset. We developed four ML models: Support Vector Machine (SVM), Random Forest, XGBoost, and Artificial Neural Network (ANN) for binary fracture outcome and 10-year fracture risk prediction. GRS and FRAX clinical risk factors (CRFs) were used as predictors. Death as a competing risk was accounted for in ML models for time-to-fracture data. ML models were subsequently fine-tuned through Bayesian optimization, which displayed marked superiority over traditional grid search. Evaluation of the models' performance considered an array of metrics such as accuracy, weighted F1 Score, the area under the precision-recall curve (PRAUC), and the area under the receiver operating characteristic curve (AUC) for binary fracture predictions, and the C-index, Brier score, and dynamic mean AUC over a 10-year follow-up period for fracture risk predictions. We found that GRS-integrated XGBoost with Bayesian optimization is the most effective model, with an accuracy of 91.2% (95% CI: 90.4-92.0%) and an AUC of 0.739 (95% CI: 0.731-0.746) in MOF binary predictions. For 10-year fracture risk modeling, the XGBoost model attained a C-index of 0.795 (95% CI: 0.783-0.806) and a mean dynamic AUC of 0.799 (95% CI: 0.788-0.809). Compared to FRAX, the XGBoost model exhibited a categorical net reclassification improvement (NRI) of 22.6% (P = .004). A sensitivity analysis, which included BMD but lacked GRS, reaffirmed these findings. Furthermore, portability tests in diverse non-European groups, including Asians and African Americans, underscored the model's robustness and adaptability. This study accentuates the potential of combining genetic insights and optimized ML in strengthening fracture predictions, heralding new preventive strategies for postmenopausal women.
Collapse
Affiliation(s)
- Qing Wu
- Department of Biomedical Informatics (Dr. Qing Wu, Jingyuan Dai), College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - Jingyuan Dai
- Department of Biomedical Informatics (Dr. Qing Wu, Jingyuan Dai), College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| |
Collapse
|
31
|
Farhat H, Makhlouf A, Gangaram P, Aifa KE, Khenissi MC, Howland I, Abid C, Jones A, Howard I, Castle N, Al Shaikh L, Khadhraoui M, Gargouri I, Laughton J, Alinier G. Exploring factors influencing time from dispatch to unit availability according to the transport decision in the pre-hospital setting: an exploratory study. BMC Emerg Med 2024; 24:77. [PMID: 38684980 PMCID: PMC11057082 DOI: 10.1186/s12873-024-00992-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/19/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Efficient resource distribution is important. Despite extensive research on response timings within ambulance services, nuances of time from unit dispatch to becoming available still need to be explored. This study aimed to identify the determinants of the duration between ambulance dispatch and readiness to respond to the next case according to the patients' transport decisions. METHODS Time from ambulance dispatch to availability (TDA) analysis according to the patients' transport decision (Transport versus Non-Transport) was conducted using R-Studio™ for a data set of 93,712 emergency calls managed by a Middle Eastern ambulance service from January to May 2023. Log-transformed Hazard Ratios (HR) were examined across diverse parameters. A Cox regression model was utilised to determine the influence of variables on TDA. Kaplan-Meier curves discerned potential variances in the time elapsed for both cohorts based on demographics and clinical indicators. A competing risk analysis assessed the probabilities of distinct outcomes occurring. RESULTS The median duration of elapsed TDA was 173 min for the transported patients and 73 min for those not transported. The HR unveiled Significant associations in various demographic variables. The Kaplan-Meier curves revealed variances in TDA across different nationalities and age categories. In the competing risk analysis, the 'Not Transported' group demonstrated a higher incidence of prolonged TDA than the 'Transported' group at specified time points. CONCLUSIONS Exploring TDA offers a novel perspective on ambulance services' efficiency. Though promising, the findings necessitate further exploration across diverse settings, ensuring broader applicability. Future research should consider a comprehensive range of variables to fully harness the utility of this period as a metric for healthcare excellence.
Collapse
Affiliation(s)
- Hassan Farhat
- Ambulance Service, Hamad Medical Corporation, PO Box 3050, Doha, Qatar.
- Faculty of Sciences, University of Sfax, 3000, Sfax, Tunisia.
- Faculty of Medicine 'Ibn El Jazzar', University of Sousse, 4000, Sousse, Tunisia.
| | - Ahmed Makhlouf
- Ambulance Service, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
- College of Engineering, Qatar University, Doha, Qatar
| | - Padarath Gangaram
- Ambulance Service, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
- Faculty of Health Sciences, Durban University of Technology, PO Box 1334, Durban, 4000, South Africa
| | - Kawther El Aifa
- Ambulance Service, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
| | | | - Ian Howland
- Ambulance Service, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
| | - Cyrine Abid
- Laboratory of Screening Cellular and Molecular Process, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Andre Jones
- Ambulance Service, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
| | - Ian Howard
- Ambulance Service, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
| | - Nicholas Castle
- Ambulance Service, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
| | - Loua Al Shaikh
- Ambulance Service, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
| | - Moncef Khadhraoui
- Higher Institute of Biotechnology, University of Sfax, Sfax, Tunisia
| | - Imed Gargouri
- Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - James Laughton
- Ambulance Service, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
| | - Guillaume Alinier
- Ambulance Service, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
- University of Hertfordshire, Hatfield, UK
- Weill Cornell Medicine-Qatar, Doha, Qatar
- Northumbria University, Newcastle Upon Tyne, UK
| |
Collapse
|
32
|
Ling Y, Liu Z, Xue JH. Survival Analysis of High-Dimensional Data With Graph Convolutional Networks and Geometric Graphs. IEEE TRANSACTIONS ON NEURAL NETWORKS AND LEARNING SYSTEMS 2024; 35:4876-4886. [PMID: 35862325 DOI: 10.1109/tnnls.2022.3190321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
This article proposes a survival model based on graph convolutional networks (GCNs) with geometric graphs directly constructed from high-dimensional features. First, we clarify that the graphs used in GCNs play an important role in processing the relational information of samples, and the graphs that align well with the underlying data structure could be beneficial for survival analysis. Second, we show that sparse geometric graphs derived from high-dimensional data are more favorable compared with dense graphs when used in GCNs for survival analysis. Third, from this insight, we propose a model for survival analysis based on GCNs. By using multiple sparse geometric graphs and a proposed sequential forward floating selection algorithm, the new model is able to simultaneously perform survival analysis and unveil the local neighborhoods of samples. The experimental results on real-world datasets show that the proposed survival analysis approach based on GCNs outperforms a variety of existing methods and indicate that geometric graphs can aid survival analysis of high-dimensional data.
Collapse
|
33
|
Li Z, Lan L, Zhou Y, Li R, Chavin KD, Xu H, Li L, Shih DJH, Jim Zheng W. Developing deep learning-based strategies to predict the risk of hepatocellular carcinoma among patients with nonalcoholic fatty liver disease from electronic health records. J Biomed Inform 2024; 152:104626. [PMID: 38521180 DOI: 10.1016/j.jbi.2024.104626] [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: 11/20/2023] [Revised: 02/23/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
OBJECTIVE The accuracy of deep learning models for many disease prediction problems is affected by time-varying covariates, rare incidence, covariate imbalance and delayed diagnosis when using structured electronic health records data. The situation is further exasperated when predicting the risk of one disease on condition of another disease, such as the hepatocellular carcinoma risk among patients with nonalcoholic fatty liver disease due to slow, chronic progression, the scarce of data with both disease conditions and the sex bias of the diseases. The goal of this study is to investigate the extent to which the aforementioned issues influence deep learning performance, and then devised strategies to tackle these challenges. These strategies were applied to improve hepatocellular carcinoma risk prediction among patients with nonalcoholic fatty liver disease. METHODS We evaluated two representative deep learning models in the task of predicting the occurrence of hepatocellular carcinoma in a cohort of patients with nonalcoholic fatty liver disease (n = 220,838) from a national EHR database. The disease prediction task was carefully formulated as a classification problem while taking censorship and the length of follow-up into consideration. RESULTS We developed a novel backward masking scheme to deal with the issue of delayed diagnosis which is very common in EHR data analysis and evaluate how the length of longitudinal information after the index date affects disease prediction. We observed that modeling time-varying covariates improved the performance of the algorithms and transfer learning mitigated reduced performance caused by the lack of data. In addition, covariate imbalance, such as sex bias in data impaired performance. Deep learning models trained on one sex and evaluated in the other sex showed reduced performance, indicating the importance of assessing covariate imbalance while preparing data for model training. CONCLUSIONS The strategies developed in this work can significantly improve the performance of hepatocellular carcinoma risk prediction among patients with nonalcoholic fatty liver disease. Furthermore, our novel strategies can be generalized to apply to other disease risk predictions using structured electronic health records, especially for disease risks on condition of another disease.
Collapse
Affiliation(s)
- Zhao Li
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - Lan Lan
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - Yujia Zhou
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - Ruoxing Li
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA
| | - Kenneth D Chavin
- Department of Surgery, Case Western Reserve University School of Medicine, 11100 Euclid Ave, Cleveland, OH 44106, USA
| | - Hua Xu
- Yale School of Medicine, USA
| | - Liang Li
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT4.6008, Houston, TX 77030, USA
| | - David J H Shih
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong Special Administrative Region
| | - W Jim Zheng
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin Street, Suite 600, Houston, TX 77030, USA.
| |
Collapse
|
34
|
Rizopoulos D, Taylor JMG. Optimizing dynamic predictions from joint models using super learning. Stat Med 2024; 43:1315-1328. [PMID: 38270062 PMCID: PMC11300862 DOI: 10.1002/sim.10010] [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: 09/20/2023] [Revised: 11/30/2023] [Accepted: 12/29/2023] [Indexed: 01/26/2024]
Abstract
Joint models for longitudinal and time-to-event data are often employed to calculate dynamic individualized predictions used in numerous applications of precision medicine. Two components of joint models that influence the accuracy of these predictions are the shape of the longitudinal trajectories and the functional form linking the longitudinal outcome history to the hazard of the event. Finding a single well-specified model that produces accurate predictions for all subjects and follow-up times can be challenging, especially when considering multiple longitudinal outcomes. In this work, we use the concept of super learning and avoid selecting a single model. In particular, we specify a weighted combination of the dynamic predictions calculated from a library of joint models with different specifications. The weights are selected to optimize a predictive accuracy metric using V-fold cross-validation. We use as predictive accuracy measures the expected quadratic prediction error and the expected predictive cross-entropy. In a simulation study, we found that the super learning approach produces results very similar to the Oracle model, which was the model with the best performance in the test datasets. All proposed methodology is implemented in the freely available R package JMbayes2.
Collapse
Affiliation(s)
- Dimitris Rizopoulos
- Department of Biostatistics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Jeremy M. G. Taylor
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
35
|
Butner JD, Dogra P, Chung C, Koay EJ, Welsh JW, Hong DS, Cristini V, Wang Z. Hybridizing mechanistic mathematical modeling with deep learning methods to predict individual cancer patient survival after immune checkpoint inhibitor therapy. RESEARCH SQUARE 2024:rs.3.rs-4151883. [PMID: 38586046 PMCID: PMC10996814 DOI: 10.21203/rs.3.rs-4151883/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
We present a study where predictive mechanistic modeling is used in combination with deep learning methods to predict individual patient survival probabilities under immune checkpoint inhibitor (ICI) therapy. This hybrid approach enables prediction based on both measures that are calculable from mechanistic models (but may not be directly measurable in the clinic) and easily measurable quantities or characteristics (that are not always readily incorporated into predictive mechanistic models). The mechanistic model we have applied here can predict tumor response from CT or MRI imaging based on key mechanisms underlying checkpoint inhibitor therapy, and in the present work, its parameters were combined with readily-available clinical measures from 93 patients into a hybrid training set for a deep learning time-to-event predictive model. Analysis revealed that training an artificial neural network with both mechanistic modeling-derived and clinical measures achieved higher per-patient predictive accuracy based on event-time concordance, Brier score, and negative binomial log-likelihood-based criteria than when only mechanistic model-derived values or only clinical data were used. Feature importance analysis revealed that both clinical and model-derived parameters play prominent roles in neural network decision making, and in increasing prediction accuracy, further supporting the advantage of our hybrid approach. We anticipate that many existing mechanistic models may be hybridized with deep learning methods in a similar manner to improve predictive accuracy through addition of additional data that may not be readily implemented in mechanistic descriptions.
Collapse
Affiliation(s)
- Joseph D Butner
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Master in Clinical Translation Management Program, The Cameron School of Business, University of St. Thomas, Houston, TX 77006, USA
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Eugene J Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas 77230, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX 77030, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medical Education, Texas A&M University School of Medicine, Bryan, TX 77807, USA
| |
Collapse
|
36
|
Lee JO, Ahn SS, Choi KS, Lee J, Jang J, Park JH, Hwang I, Park CK, Park SH, Chung JW, Choi SH. Added prognostic value of 3D deep learning-derived features from preoperative MRI for adult-type diffuse gliomas. Neuro Oncol 2024; 26:571-580. [PMID: 37855826 PMCID: PMC10912011 DOI: 10.1093/neuonc/noad202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND To investigate the prognostic value of spatial features from whole-brain MRI using a three-dimensional (3D) convolutional neural network for adult-type diffuse gliomas. METHODS In a retrospective, multicenter study, 1925 diffuse glioma patients were enrolled from 5 datasets: SNUH (n = 708), UPenn (n = 425), UCSF (n = 500), TCGA (n = 160), and Severance (n = 132). The SNUH and Severance datasets served as external test sets. Precontrast and postcontrast 3D T1-weighted, T2-weighted, and T2-FLAIR images were processed as multichannel 3D images. A 3D-adapted SE-ResNeXt model was trained to predict overall survival. The prognostic value of the deep learning-based prognostic index (DPI), a spatial feature-derived quantitative score, and established prognostic markers were evaluated using Cox regression. Model evaluation was performed using the concordance index (C-index) and Brier score. RESULTS The MRI-only median DPI survival prediction model achieved C-indices of 0.709 and 0.677 (BS = 0.142 and 0.215) and survival differences (P < 0.001 and P = 0.002; log-rank test) for the SNUH and Severance datasets, respectively. Multivariate Cox analysis revealed DPI as a significant prognostic factor, independent of clinical and molecular genetic variables: hazard ratio = 0.032 and 0.036 (P < 0.001 and P = 0.004) for the SNUH and Severance datasets, respectively. Multimodal prediction models achieved higher C-indices than models using only clinical and molecular genetic variables: 0.783 vs. 0.774, P = 0.001, SNUH; 0.766 vs. 0.748, P = 0.023, Severance. CONCLUSIONS The global morphologic feature derived from 3D CNN models using whole-brain MRI has independent prognostic value for diffuse gliomas. Combining clinical, molecular genetic, and imaging data yields the best performance.
Collapse
Affiliation(s)
- Jung Oh Lee
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Artificial Intelligence Collaborative Network, Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung Soo Ahn
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyu Sung Choi
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Artificial Intelligence Collaborative Network, Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Junhyeok Lee
- Interdisciplinary Programs in Cancer Biology Major, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Joon Jang
- Department of Biomedical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jung Hyun Park
- Department of Radiology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Inpyeong Hwang
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Artificial Intelligence Collaborative Network, Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung Hye Park
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jin Wook Chung
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Artificial Intelligence Collaborative Network, Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Innovate Biomedical Technology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung Hong Choi
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Artificial Intelligence Collaborative Network, Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea
| |
Collapse
|
37
|
Sun Y, Hu S, Li X, Wu Y. Development and Application of a Novel Machine Learning Model Predicting Pancreatic Cancer-Specific Mortality. Cureus 2024; 16:e57161. [PMID: 38681451 PMCID: PMC11056009 DOI: 10.7759/cureus.57161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2024] [Indexed: 05/01/2024] Open
Abstract
Precise prognostication is vital for guiding treatment decisions in people diagnosed with pancreatic cancer. Existing models depend on predetermined variables, constraining their effectiveness. Our objective was to explore a novel machine learning approach to enhance a prognostic model for predicting pancreatic cancer-specific mortality and, subsequently, to assess its performance against Cox regression models. Datasets were retrospectively collected and analyzed for 9,752 patients diagnosed with pancreatic cancer and with surgery performed. The primary outcomes were the mortality of patients with pancreatic carcinoma at one year, three years, and five years. Model discrimination was assessed using the concordance index (C-index), and calibration was assessed using Brier scores. The Survival Quilts model was compared with Cox regression models in clinical use, and decision curve analysis was done. The Survival Quilts model demonstrated robust discrimination for one-year (C-index 0.729), three-year (C-index 0.693), and five-year (C-index 0.672) pancreatic cancer-specific mortality. In comparison to Cox models, the Survival Quilts models exhibited a higher C-index up to 32 months but displayed inferior performance after 33 months. A subgroup analysis was conducted, revealing that within the subset of individuals without metastasis, the Survival Quilts models showcased a significant advantage over the Cox models. In the cohort with metastatic pancreatic cancer, Survival Quilts outperformed the Cox model before 24 months but exhibited a weaker performance after 25 months. This study has developed and validated a novel machine learning-based Survival Quilts model to predict pancreatic cancer-specific mortality that outperforms the Cox regression model.
Collapse
Affiliation(s)
- Yongji Sun
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, CHN
| | - Sien Hu
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, CHN
| | - Xiawei Li
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, CHN
| | - Yulian Wu
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, CHN
| |
Collapse
|
38
|
Nguyen H, Vasconcellos HD, Keck K, Carr J, Launer LJ, Guallar E, Lima JAC, Ambale-Venkatesh B. Utility of multimodal longitudinal imaging data for dynamic prediction of cardiovascular and renal disease: the CARDIA study. FRONTIERS IN RADIOLOGY 2024; 4:1269023. [PMID: 38476649 PMCID: PMC10927728 DOI: 10.3389/fradi.2024.1269023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 02/06/2024] [Indexed: 03/14/2024]
Abstract
Background Medical examinations contain repeatedly measured data from multiple visits, including imaging variables collected from different modalities. However, the utility of such data for the prediction of time-to-event is unknown, and only a fraction of the data is typically used for risk prediction. We hypothesized that multimodal longitudinal imaging data could improve dynamic disease prognosis of cardiovascular and renal disease (CVRD). Methods In a multi-centered cohort of 5,114 CARDIA participants, we included 166 longitudinal imaging variables from five imaging modalities: Echocardiography (Echo), Cardiac and Abdominal Computed Tomography (CT), Dual-Energy x-ray Absorptiometry (DEXA), Brain Magnetic Resonance Imaging (MRI) collected from young adulthood to mid-life over 30 years (1985-2016) to perform dynamic survival analysis of CVRD events using machine learning dynamic survival analysis (Dynamic-DeepHit, LTRCforest, and Extended Cox for Time-varying Covariates). Risk probabilities were continuously updated as new data were collected. Model performance was assessed using integrated AUC and C-index and compared to traditional risk factors. Results Longitudinal imaging data, even when being irregularly collected with high missing rates, improved CVRD dynamic prediction (0.03 in integrated AUC, up to 0.05 in C-index compared to traditional risk factors; best model's C-index = 0.80-0.83 up to 20 years from baseline) from young adulthood followed up to midlife. Among imaging variables, Echo and CT variables contributed significantly to improved risk estimation. Echo measured in early adulthood predicted midlife CVRD risks almost as well as Echo measured 10-15 years later (0.01 C-index difference). The most recent CT exam provided the most accurate prediction for short-term risk estimation. Brain MRI markers provided additional information from cardiac Echo and CT variables that led to a slightly improved prediction. Conclusions Longitudinal multimodal imaging data readily collected from follow-up exams can improve CVRD dynamic prediction. Echocardiography measured early can provide a good long-term risk estimation, while CT/calcium scoring variables carry atherosclerotic signatures that benefit more immediate risk assessment starting in middle-age.
Collapse
Affiliation(s)
- Hieu Nguyen
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | | | - Kimberley Keck
- Department of Cardiology, Johns Hopkins University, Baltimore, MD, United States
| | - Jeffrey Carr
- Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, TN, United States
| | - Lenore J. Launer
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Bethesda, MD, United States
| | - Eliseo Guallar
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - João A. C. Lima
- Department of Cardiology, Johns Hopkins University, Baltimore, MD, United States
| | | |
Collapse
|
39
|
Yu J, Yang X, Deng Y, Krefman AE, Pool LR, Zhao L, Mi X, Ning H, Wilkins J, Lloyd-Jones DM, Petito LC, Allen NB. Incorporating longitudinal history of risk factors into atherosclerotic cardiovascular disease risk prediction using deep learning. Sci Rep 2024; 14:2554. [PMID: 38296982 PMCID: PMC10830564 DOI: 10.1038/s41598-024-51685-5] [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: 10/02/2023] [Accepted: 01/08/2024] [Indexed: 02/02/2024] Open
Abstract
It is increasingly clear that longitudinal risk factor levels and trajectories are related to risk for atherosclerotic cardiovascular disease (ASCVD) above and beyond single measures. Currently used in clinical care, the Pooled Cohort Equations (PCE) are based on regression methods that predict ASCVD risk based on cross-sectional risk factor levels. Deep learning (DL) models have been developed to incorporate longitudinal data for risk prediction but its benefit for ASCVD risk prediction relative to the traditional Pooled Cohort Equations (PCE) remain unknown. Our study included 15,565 participants from four cardiovascular disease cohorts free of baseline ASCVD who were followed for adjudicated ASCVD. Ten-year ASCVD risk was calculated in the training set using our benchmark, the PCE, and a longitudinal DL model, Dynamic-DeepHit. Predictors included those incorporated in the PCE: sex, race, age, total cholesterol, high density lipid cholesterol, systolic and diastolic blood pressure, diabetes, hypertension treatment and smoking. The discrimination and calibration performance of the two models were evaluated in an overall hold-out testing dataset. Of the 15,565 participants in our dataset, 2170 (13.9%) developed ASCVD. The performance of the longitudinal DL model that incorporated 8 years of longitudinal risk factor data improved upon that of the PCE [AUROC: 0.815 (CI 0.782-0.844) vs 0.792 (CI 0.760-0.825)] and the net reclassification index was 0.385. The brier score for the DL model was 0.0514 compared with 0.0542 in the PCE. Incorporating longitudinal risk factors in ASCVD risk prediction using DL can improve model discrimination and calibration.
Collapse
Affiliation(s)
- Jingzhi Yu
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Xiaoyun Yang
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yu Deng
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Amy E Krefman
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lindsay R Pool
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lihui Zhao
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Xinlei Mi
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hongyan Ning
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - John Wilkins
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Donald M Lloyd-Jones
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lucia C Petito
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Norrina B Allen
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
40
|
Mahootiha M, Qadir HA, Aghayan D, Fretland ÅA, von Gohren Edwin B, Balasingham I. Deep learning-assisted survival prognosis in renal cancer: A CT scan-based personalized approach. Heliyon 2024; 10:e24374. [PMID: 38298725 PMCID: PMC10828686 DOI: 10.1016/j.heliyon.2024.e24374] [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: 07/17/2023] [Revised: 12/19/2023] [Accepted: 01/08/2024] [Indexed: 02/02/2024] Open
Abstract
This paper presents a deep learning (DL) approach for predicting survival probabilities of renal cancer patients based solely on preoperative CT imaging. The proposed approach consists of two networks: a classifier- and a survival- network. The classifier attempts to extract features from 3D CT scans to predict the ISUP grade of Renal cell carcinoma (RCC) tumors, as defined by the International Society of Urological Pathology (ISUP). Our classifier is a 3D convolutional neural network to avoid losing crucial information on the interconnection of slides in 3D images. We employ multiple procedures, including image augmentation, preprocessing, and concatenation, to improve the performance of the classifier. Given the strong correlation between ISUP grading and renal cancer prognosis in the clinical context, we use the ISUP grading features extracted by the classifier as the input to the survival network. By leveraging this clinical association and the classifier network, we are able to model our survival analysis using a simple DL-based network. We adopt a discrete LogisticHazard-based loss to extract intrinsic survival characteristics of RCC tumors from CT images. This allows us to build a completely parametric survival model that varies with patients' tumor characteristics and predicts non-proportional survival probability curves for different patients. Our results demonstrated that the proposed method could predict the future course of renal cancer with reasonable accuracy from the CT scans. The proposed method obtained an average concordance index of 0.72, an integrated Brier score of 0.15, and an area under the curve value of 0.71 on the test cohorts.
Collapse
Affiliation(s)
- Maryamalsadat Mahootiha
- The Intervention Centre, Oslo University Hospital, Oslo, 0372, Norway
- Faculty of Medicine, University of Oslo, Oslo, 0372, Norway
| | - Hemin Ali Qadir
- The Intervention Centre, Oslo University Hospital, Oslo, 0372, Norway
| | - Davit Aghayan
- The Intervention Centre, Oslo University Hospital, Oslo, 0372, Norway
| | | | - Bjørn von Gohren Edwin
- The Intervention Centre, Oslo University Hospital, Oslo, 0372, Norway
- Faculty of Medicine, University of Oslo, Oslo, 0372, Norway
| | - Ilangko Balasingham
- The Intervention Centre, Oslo University Hospital, Oslo, 0372, Norway
- Department of Electronic Systems, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
41
|
Kolk MZH, Ruipérez-Campillo S, Alvarez-Florez L, Deb B, Bekkers EJ, Allaart CP, Van Der Lingen ALCJ, Clopton P, Išgum I, Wilde AAM, Knops RE, Narayan SM, Tjong FVY. Dynamic prediction of malignant ventricular arrhythmias using neural networks in patients with an implantable cardioverter-defibrillator. EBioMedicine 2024; 99:104937. [PMID: 38118401 PMCID: PMC10772563 DOI: 10.1016/j.ebiom.2023.104937] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/20/2023] [Accepted: 12/12/2023] [Indexed: 12/22/2023] Open
Abstract
BACKGROUND Risk stratification for ventricular arrhythmias currently relies on static measurements that fail to adequately capture dynamic interactions between arrhythmic substrate and triggers over time. We trained and internally validated a dynamic machine learning (ML) model and neural network that extracted features from longitudinally collected electrocardiograms (ECG), and used these to predict the risk of malignant ventricular arrhythmias. METHODS A multicentre study in patients implanted with an implantable cardioverter-defibrillator (ICD) between 2007 and 2021 in two academic hospitals was performed. Variational autoencoders (VAEs), which combine neural networks with variational inference principles, and can learn patterns and structure in data without explicit labelling, were trained to encode the mean ECG waveforms from the limb leads into 16 variables. Supervised dynamic ML models using these latent ECG representations and clinical baseline information were trained to predict malignant ventricular arrhythmias treated by the ICD. Model performance was evaluated on a hold-out set, using time-dependent receiver operating characteristic (ROC) and calibration curves. FINDINGS 2942 patients (61.7 ± 13.9 years, 25.5% female) were included, with a total of 32,129 ECG recordings during a mean follow-up of 43.9 ± 35.9 months. The mean time-varying area under the ROC curve for the dynamic model was 0.738 ± 0.07, compared to 0.639 ± 0.03 for a static (i.e. baseline-only model). Feature analyses indicated dynamic changes in latent ECG representations, particularly those affecting the T-wave morphology, were of highest importance for model predictions. INTERPRETATION Dynamic ML models and neural networks effectively leverage routinely collected longitudinal ECG recordings for personalised and updated predictions of malignant ventricular arrhythmias, outperforming static models. FUNDING This publication is part of the project DEEP RISK ICD (with project number 452019308) of the research programme Rubicon which is (partly) financed by the Dutch Research Council (NWO). This research is partly funded by the Amsterdam Cardiovascular Sciences (personal grant F.V.Y.T).
Collapse
Affiliation(s)
- Maarten Z H Kolk
- Department of Clinical and Experimental Cardiology, Amsterdam UMC Location University of Amsterdam, Heart Center, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Samuel Ruipérez-Campillo
- Department of Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, USA; Department of Information Technology and Electrical Engineering, Swiss Federal Institute of Technology Zurich (ETHz), Gloriastrasse 35, Zurich, Switzerland; ITACA Institute, Universtitat Politècnica de València, Camino de Vera S/n, Valencia, Spain
| | - Laura Alvarez-Florez
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Center Location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Brototo Deb
- Department of Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Erik J Bekkers
- Faculty of Science, University of Amsterdam, Science Park 904, Amsterdam, the Netherlands
| | - Cornelis P Allaart
- Department of Cardiology, Amsterdam UMC, Location VU Medical Center, De Boelelaan 1118, Amsterdam, the Netherlands
| | | | - Paul Clopton
- Department of Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Ivana Išgum
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Center Location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Faculty of Science, University of Amsterdam, Science Park 904, Amsterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Arthur A M Wilde
- Department of Clinical and Experimental Cardiology, Amsterdam UMC Location University of Amsterdam, Heart Center, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Reinoud E Knops
- Department of Clinical and Experimental Cardiology, Amsterdam UMC Location University of Amsterdam, Heart Center, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Sanjiv M Narayan
- Department of Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Fleur V Y Tjong
- Department of Clinical and Experimental Cardiology, Amsterdam UMC Location University of Amsterdam, Heart Center, Meibergdreef 9, Amsterdam, the Netherlands; Department of Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, USA; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands.
| |
Collapse
|
42
|
Zhao Z, Li W, Liu P, Zhang A, Sun J, Xu LX. Survival Analysis for Multimode Ablation Using Self-Adapted Deep Learning Network Based on Multisource Features. IEEE J Biomed Health Inform 2024; 28:19-30. [PMID: 37015120 DOI: 10.1109/jbhi.2023.3260776] [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: 04/03/2023]
Abstract
Novel multimode thermal therapy by freezing before radio-frequency heating has achieved a desirable therapeutic effect in liver cancer. Compared with surgical resection, ablation treatment has a relatively high risk of tumor recurrence. To monitor tumor progression after ablation, we developed a novel survival analysis framework for survival prediction and efficacy assessment. We extracted preoperative and postoperative MRI radiomics features and vision transformer-based deep learning features. We also combined the immune features extracted from peripheral blood immune responses using flow cytometry and routine blood tests before and after treatment. We selected features using random survival forest and improved the deep Cox mixture (DCM) for survival analysis. To properly accommodate multitype input features, we proposed a self-adapted fully connected layer for locally and globally representing features. We evaluated the method using our clinical dataset. Of note, the immune features rank the highest feature importance and contribute significantly to the prediction accuracy. The results showed a promising C$^{\mathit{td}}$-index of 0.885 $\pm$ 0.040 and an integrated Brier score of 0.041 $\pm$ 0.014, which outperformed state-of-the-art method combinations of survival prediction. For each patient, individual survival probability was accurately predicted over time, which provided clinicians with trustable prognosis suggestions.
Collapse
|
43
|
Infante G, Miceli R, Ambrogi F. Sample size and predictive performance of machine learning methods with survival data: A simulation study. Stat Med 2023; 42:5657-5675. [PMID: 37947168 DOI: 10.1002/sim.9931] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 09/11/2023] [Accepted: 09/21/2023] [Indexed: 11/12/2023]
Abstract
Prediction models are increasingly developed and used in diagnostic and prognostic studies, where the use of machine learning (ML) methods is becoming more and more popular over traditional regression techniques. For survival outcomes the Cox proportional hazards model is generally used and it has been proven to achieve good prediction performances with few strong covariates. The possibility to improve the model performance by including nonlinearities, covariate interactions and time-varying effects while controlling for overfitting must be carefully considered during the model building phase. On the other hand, ML techniques are able to learn complexities from data at the cost of hyper-parameter tuning and interpretability. One aspect of special interest is the sample size needed for developing a survival prediction model. While there is guidance when using traditional statistical models, the same does not apply when using ML techniques. This work develops a time-to-event simulation framework to evaluate performances of Cox regression compared, among others, to tuned random survival forest, gradient boosting, and neural networks at varying sample sizes. Simulations were based on replications of subjects from publicly available databases, where event times were simulated according to a Cox model with nonlinearities on continuous variables and time-varying effects and on the SEER registry data.
Collapse
Affiliation(s)
- Gabriele Infante
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Unit of Biostatistics for Clinical Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rosalba Miceli
- Unit of Biostatistics for Clinical Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federico Ambrogi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Scientific Directorate, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| |
Collapse
|
44
|
Qi SA, Kumar N, Verma R, Xu JY, Shen-Tu G, Greiner R. Using Bayesian Neural Networks to Select Features and Compute Credible Intervals for Personalized Survival Prediction. IEEE Trans Biomed Eng 2023; 70:3389-3400. [PMID: 37339045 DOI: 10.1109/tbme.2023.3287514] [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: 06/22/2023]
Abstract
An Individual Survival Distribution (ISD) models a patient's personalized survival probability at all future time points. Previously, ISD models have been shown to produce accurate and personalized survival estimates (for example, time to relapse or to death) in several clinical applications. However, off-the-shelf neural-network-based ISD models are usually opaque models due to their limited support for meaningful feature selection and uncertainty estimation, which hinders their wide clinical adoption. Here, we introduce a Bayesian-neural-network-based ISD (BNN-ISD) model that produces accurate survival estimates but also quantifies the uncertainty in model's parameter estimation, which can be used to (1) rank the importance of the input features to support feature selection and (2) compute credible intervals around ISDs for clinicians to assess the model's confidence in its prediction. Our BNN-ISD model utilized sparsity-inducing priors to learn a sparse set of weights to enable feature selection. We provide empirical evidence, on 2 synthetic and 3 real-world clinical datasets, that BNN-ISD system can effectively select meaningful features and compute trustworthy credible intervals of the survival distribution for each patient. We observed that our approach accurately recovers feature importance in the synthetic datasets and selects meaningful features for the real-world clinical data as well, while also achieving state-of-the-art survival prediction performance. We also show that these credible regions can aid in clinical decision-making by providing a gauge of the uncertainty of the estimated ISD curves.
Collapse
|
45
|
Bonomi L, Lionts M, Fan L. Private Continuous Survival Analysis with Distributed Multi-Site Data. PROCEEDINGS : ... IEEE INTERNATIONAL CONFERENCE ON BIG DATA. IEEE INTERNATIONAL CONFERENCE ON BIG DATA 2023; 2023:5444-5453. [PMID: 38585488 PMCID: PMC10997374 DOI: 10.1109/bigdata59044.2023.10386571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Effective disease surveillance systems require large-scale epidemiological data to improve health outcomes and quality of care for the general population. As data may be limited within a single site, multi-site data (e.g., from a number of local/regional health systems) need to be considered. Leveraging distributed data across multiple sites for epidemiological analysis poses significant challenges. Due to the sensitive nature of epidemiological data, it is imperative to design distributed solutions that provide strong privacy protections. Current privacy solutions often assume a central site, which is responsible for aggregating the distributed data and applying privacy protection before sharing the results (e.g., aggregation via secure primitives and differential privacy for sharing aggregate results). However, identifying such a central site may be difficult in practice and relying on a central site may introduce potential vulnerabilities (e.g., single point of failure). Furthermore, to support clinical interventions and inform policy decisions in a timely manner, epidemiological analysis need to reflect dynamic changes in the data. Yet, existing distributed privacy-protecting approaches were largely designed for static data (e.g., one-time data sharing) and cannot fulfill dynamic data requirements. In this work, we propose a privacy-protecting approach that supports the sharing of dynamic epidemiological analysis and provides strong privacy protection in a decentralized manner. We apply our solution in continuous survival analysis using the Kaplan-Meier estimation model while providing differential privacy protection. Our evaluations on a real dataset containing COVID-19 cases show that our method provides highly usable results.
Collapse
Affiliation(s)
- Luca Bonomi
- Dept. Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Marilyn Lionts
- Dept. Computer Science, Vanderbilt University, Nashville, TN
| | - Liyue Fan
- College of Computing and Informatics, University of North Carolina, Charlotte, NC
| |
Collapse
|
46
|
Liu Y, Sun S, Zhang Y, Huang X, Wang K, Qu Y, Chen X, Wu R, Zhang J, Luo J, Li Y, Wang J, Yi J. Predictive function of tumor burden-incorporated machine-learning algorithms for overall survival and their value in guiding management decisions in patients with locally advanced nasopharyngeal carcinoma. JOURNAL OF THE NATIONAL CANCER CENTER 2023; 3:295-305. [PMID: 39036668 PMCID: PMC11256522 DOI: 10.1016/j.jncc.2023.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 07/23/2024] Open
Abstract
Objective Accurate prognostic predictions and personalized decision-making on induction chemotherapy (IC) for individuals with locally advanced nasopharyngeal carcinoma (LA-NPC) remain challenging. This research examined the predictive function of tumor burden-incorporated machine-learning algorithms for overall survival (OS) and their value in guiding treatment in patients with LA-NPC. Methods Individuals with LA-NPC were reviewed retrospectively. Tumor burden signature-based OS prediction models were established using a nomogram and two machine-learning methods, the interpretable eXtreme Gradient Boosting (XGBoost) risk prediction model, and DeepHit time-to-event neural network. The models' prediction performances were compared using the concordance index (C-index) and the area under the curve (AUC). The patients were divided into two cohorts based on the risk predictions of the most successful model. The efficacy of IC combined with concurrent chemoradiotherapy was compared to that of chemoradiotherapy alone. Results The 1 221 eligible individuals, assigned to the training (n = 813) or validation (n = 408) set, showed significant respective differences in the C-indices of the XGBoost, DeepHit, and nomogram models (0.849 and 0.768, 0.811 and 0.767, 0.730 and 0.705). The training and validation sets had larger AUCs in the XGBoost and DeepHit models than the nomogram model in predicting OS (0.881 and 0.760, 0.845 and 0.776, and 0.764 and 0.729, P < 0.001). IC presented survival benefits in the XGBoost-derived high-risk but not low-risk group. Conclusion This research used machine-learning algorithms to create and verify a comprehensive model integrating tumor burden with clinical variables to predict OS and determine which patients will most likely gain from IC. This model could be valuable for delivering patient counseling and conducting clinical evaluations.
Collapse
Affiliation(s)
- Yang Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shiran Sun
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ye Zhang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaodong Huang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Kai Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuan Qu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xuesong Chen
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Runye Wu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jianghu Zhang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jingwei Luo
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yexiong Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jingbo Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Junlin Yi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Hebei Cancer Hospital, Chinese Academy of Medical Sciences (CAMS), Langfang 065001, China
| |
Collapse
|
47
|
Zhang D, Luan J, Liu B, Yang A, Lv K, Hu P, Han X, Yu H, Shmuel A, Ma G, Zhang C. Comparison of MRI radiomics-based machine learning survival models in predicting prognosis of glioblastoma multiforme. Front Med (Lausanne) 2023; 10:1271687. [PMID: 38098850 PMCID: PMC10720716 DOI: 10.3389/fmed.2023.1271687] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/15/2023] [Indexed: 12/17/2023] Open
Abstract
OBJECTIVE To compare the performance of radiomics-based machine learning survival models in predicting the prognosis of glioblastoma multiforme (GBM) patients. METHODS 131 GBM patients were included in our study. The traditional Cox proportional-hazards (CoxPH) model and four machine learning models (SurvivalTree, Random survival forest (RSF), DeepSurv, DeepHit) were constructed, and the performance of the five models was evaluated using the C-index. RESULTS After the screening, 1792 radiomics features were obtained. Seven radiomics features with the strongest relationship with prognosis were obtained following the application of the least absolute shrinkage and selection operator (LASSO) regression. The CoxPH model demonstrated that age (HR = 1.576, p = 0.037), Karnofsky performance status (KPS) score (HR = 1.890, p = 0.006), radiomics risk score (HR = 3.497, p = 0.001), and radiomics risk level (HR = 1.572, p = 0.043) were associated with poorer prognosis. The DeepSurv model performed the best among the five models, obtaining C-index of 0.882 and 0.732 for the training and test set, respectively. The performances of the other four models were lower: CoxPH (0.663 training set / 0.635 test set), SurvivalTree (0.702/0.655), RSF (0.735/0.667), DeepHit (0.608/0.560). CONCLUSION This study confirmed the superior performance of deep learning algorithms based on radiomics relative to the traditional method in predicting the overall survival of GBM patients; specifically, the DeepSurv model showed the best predictive ability.
Collapse
Affiliation(s)
- Di Zhang
- Department of Radiology, Liaocheng People’s Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Liaocheng, Shandong, China
| | - Jixin Luan
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Bing Liu
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Aocai Yang
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Kuan Lv
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Pianpian Hu
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xiaowei Han
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Hongwei Yu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Amir Shmuel
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Chuanchen Zhang
- Department of Radiology, Liaocheng People’s Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Liaocheng, Shandong, China
| |
Collapse
|
48
|
Yang K, Zhu G, Sun Y, Hu Y, Lv Y, Li Y, Pan J, Chen F, Zhou Y, Zhang J. Prognostic significance of cyclin D1 expression pattern in HPV-negative oral and oropharyngeal carcinoma: A deep-learning approach. J Oral Pathol Med 2023; 52:919-929. [PMID: 37701976 DOI: 10.1111/jop.13482] [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: 03/27/2023] [Revised: 08/09/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND We aimed to establish image recognition and survival prediction models using a novel scoring system of cyclin D1 expression pattern in patients with human papillomavirus-negative oral or oropharyngeal squamous cell carcinoma. METHODS The clinicopathological data of 610 patients with human papillomavirus-negative oral/oropharyngeal squamous cell carcinoma were analyzed retrospectively. Cox univariate and multivariate risk regression analyses were performed to compare cyclin D1 expression pattern scoring with the traditional scoring method-cyclin D1 expression level scoring-in relation to patients' overall and progression-free survival. An image recognition model employing the cyclin D1 expression pattern scoring system was established by YOLOv5 algorithms. From this model, two independent survival prediction models were established using the DeepHit and DeepSurv algorithms. RESULTS Cyclin D1 had three expression patterns in oral and oropharyngeal squamous cell carcinoma cancer nests. Superior to cyclin D1 expression level scoring, cyclin D1 expression pattern scoring was significantly correlated with the prognosis of patients with oral squamous cell carcinoma (p < 0.0001) and oropharyngeal squamous cell carcinoma (p < 0.05). Moreover, it was an independent prognostic risk factor in both oral squamous cell carcinoma (p < 0.0001) and oropharyngeal squamous cell carcinoma (p < 0.05). The cyclin D1 expression pattern-derived image recognition model showed an average test set accuracy of 78.48% ± 4.31%. In the overall survival prediction models, the average concordance indices of the test sets established by DeepSurv and DeepHit were 0.71 ± 0.02 and 0.70 ± 0.01, respectively. CONCLUSION Combined with the image recognition model of the cyclin D1 expression pattern, the survival prediction model had a relatively good prediction effect on the overall survival prognosis of patients with human papillomavirus-negative oral or oropharyngeal squamous cell carcinoma.
Collapse
Affiliation(s)
- Ke Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guixin Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Other Research Platforms, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanan Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yaying Hu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yinan Lv
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yiwei Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Juncheng Pan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Fu Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jiali Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
49
|
Stankevičiūtė K, Woillard JB, Peck RW, Marquet P, van der Schaar M. Bridging the Worlds of Pharmacometrics and Machine Learning. Clin Pharmacokinet 2023; 62:1551-1565. [PMID: 37803104 DOI: 10.1007/s40262-023-01310-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2023] [Indexed: 10/08/2023]
Abstract
Precision medicine requires individualized modeling of disease and drug dynamics, with machine learning-based computational techniques gaining increasing popularity. The complexity of either field, however, makes current pharmacological problems opaque to machine learning practitioners, and state-of-the-art machine learning methods inaccessible to pharmacometricians. To help bridge the two worlds, we provide an introduction to current problems and techniques in pharmacometrics that ranges from pharmacokinetic and pharmacodynamic modeling to pharmacometric simulations, model-informed precision dosing, and systems pharmacology, and review some of the machine learning approaches to address them. We hope this would facilitate collaboration between experts, with complementary strengths of principled pharmacometric modeling and flexibility of machine learning leading to synergistic effects in pharmacological applications.
Collapse
Affiliation(s)
- Kamilė Stankevičiūtė
- Department of Computer Science and Technology, University of Cambridge, 15 JJ Thomson Avenue, Cambridge, CB3 0FD, UK
| | - Jean-Baptiste Woillard
- INSERM U1248 P&T, University of Limoges, 2 rue du Pr Descottes, 87000, Limoges, France.
- Department of Pharmacology and Toxicology, CHU Limoges, Limoges, France.
| | - Richard W Peck
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
- Pharma Research and Development, Roche Innovation Center, Basel, Switzerland
| | - Pierre Marquet
- INSERM U1248 P&T, University of Limoges, 2 rue du Pr Descottes, 87000, Limoges, France
- Department of Pharmacology and Toxicology, CHU Limoges, Limoges, France
| | - Mihaela van der Schaar
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, UK
- The Alan Turing Institute, London, UK
| |
Collapse
|
50
|
Yu J, Yang X, Deng Y, Krefman AE, Pool LR, Zhao L, Mi X, Ning H, Wilkins J, Lloyd-Jones DM, Petito LC, Allen NB. Incorporating longitudinal history of risk factors into atherosclerotic cardiovascular disease risk prediction using deep learning. RESEARCH SQUARE 2023:rs.3.rs-3405388. [PMID: 37886463 PMCID: PMC10602136 DOI: 10.21203/rs.3.rs-3405388/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Background It is increasingly clear that longitudinal risk factor levels and trajectories are related to risk for atherosclerotic cardiovascular disease (ASCVD) above and beyond single measures. Currently used in clinical care, the Pooled Cohort Equations (PCE) are based on regression methods that predict ASCVD risk based on cross-sectional risk factor levels. Deep learning (DL) models have been developed to incorporate longitudinal data for risk prediction but its benefit for ASCVD risk prediction relative to the traditional Pooled Cohort Equations (PCE) remain unknown. Objective To develop a ASCVD risk prediction model that incorporates longitudinal risk factors using deep learning. Methods Our study included 15,565 participants from four cardiovascular disease cohorts free of baseline ASCVD who were followed for adjudicated ASCVD. Ten-year ASCVD risk was calculated in the training set using our benchmark, the PCE, and a longitudinal DL model, Dynamic-DeepHit. Predictors included those incorporated in the PCE: sex, race, age, total cholesterol, high density lipid cholesterol, systolic and diastolic blood pressure, diabetes, hypertension treatment and smoking. The discrimination and calibration performance of the two models were evaluated in an overall hold-out testing dataset. Results Of the 15,565 participants in our dataset, 2,170 (13.9%) developed ASCVD. The performance of the longitudinal DL model that incorporated 8 years of longitudinal risk factor data improved upon that of the PCE [AUROC: 0.815 (CI: 0.782-0.844) vs 0.792 (CI: 0.760-0.825)] and the net reclassification index was 0.385. The brier score for the DL model was 0.0514 compared with 0.0542 in the PCE. Conclusion Incorporating longitudinal risk factors in ASCVD risk prediction using DL can improve model discrimination and calibration.
Collapse
|