Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 398
Filtrar
1.
Biomed Tech (Berl) ; 2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-39241784

RESUMO

In this paper, the multi-task dense-feature-fusion survival prediction (DFFSP) model is proposed to predict the three-year survival for glioblastoma (GBM) patients based on radiogenomics data. The contrast-enhanced T1-weighted (T1w) image, T2-weighted (T2w) image and copy number variation (CNV) is used as the input of the three branches of the DFFSP model. This model uses two image extraction modules consisting of residual blocks and one dense feature fusion module to make multi-scale fusion of T1w and T2w image features as backbone. Also, a gene feature extraction module is used to adaptively weight CNV fragments. Besides, a transfer learning module is introduced to solve the small sample problem and an image reconstruction module is adopted to make the model anatomy-aware under a multi-task framework. 256 sample pairs (T1w and corresponding T2w MRI slices) and 187 CNVs of 74 patients were used. The experimental results show that the proposed model can predict the three-year survival of GBM patients with the accuracy of 89.1 %, which is improved by 3.2 and 4.7 % compared with the model without genes and the model using last fusion strategy, respectively. This model could also classify the patients into high-risk and low-risk groups, which will effectively assist doctors in diagnosing GBM patients.

2.
Neurooncol Adv ; 6(1): vdae122, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39156618

RESUMO

Background: This research aims to improve glioblastoma survival prediction by integrating MR images, clinical, and molecular-pathologic data in a transformer-based deep learning model, addressing data heterogeneity and performance generalizability. Methods: We propose and evaluate a transformer-based nonlinear and nonproportional survival prediction model. The model employs self-supervised learning techniques to effectively encode the high-dimensional MRI input for integration with nonimaging data using cross-attention. To demonstrate model generalizability, the model is assessed with the time-dependent concordance index (Cdt) in 2 training setups using 3 independent public test sets: UPenn-GBM, UCSF-PDGM, and Rio Hortega University Hospital (RHUH)-GBM, each comprising 378, 366, and 36 cases, respectively. Results: The proposed transformer model achieved a promising performance for imaging as well as nonimaging data, effectively integrating both modalities for enhanced performance (UCSF-PDGM test-set, imaging Cdt 0.578, multimodal Cdt 0.672) while outperforming state-of-the-art late-fusion 3D-CNN-based models. Consistent performance was observed across the 3 independent multicenter test sets with Cdt values of 0.707 (UPenn-GBM, internal test set), 0.672 (UCSF-PDGM, first external test set), and 0.618 (RHUH-GBM, second external test set). The model achieved significant discrimination between patients with favorable and unfavorable survival for all 3 datasets (log-rank P 1.9 × 10-8, 9.7 × 10-3, and 1.2 × 10-2). Comparable results were obtained in the second setup using UCSF-PDGM for training/internal testing and UPenn-GBM and RHUH-GBM for external testing (Cdt 0.670, 0.638, and 0.621). Conclusions: The proposed transformer-based survival prediction model integrates complementary information from diverse input modalities, contributing to improved glioblastoma survival prediction compared to state-of-the-art methods. Consistent performance was observed across institutions supporting model generalizability.

3.
Int J Mol Cell Med ; 13(1): 79-90, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39156868

RESUMO

Glioblastoma (GBM) is the most aggressive and lethal brain tumor. Artificial neural networks (ANNs) have the potential to make accurate predictions and improve decision making. The aim of this study was to create an ANN model to predict 15-month survival in GBM patients according to gene expression databases. Genomic data of GBM were downloaded from the CGGA, TCGA, MYO, and CPTAC. Logistic regression (LR) and ANN model were used. Age, gender, IDH wild-type/mutant and the 31 most important genes from our previous study, were determined as input factors for the established ANN model. 15-month survival time was used to evaluate the results. The normalized importance scores of each covariate were calculated using the selected ANN model. The area under a receiver operating characteristic (ROC) curve (AUC), Hosmer-Lemeshow (H-L) statistic and accuracy of prediction were measured to evaluate the two models. SPSS 26 was utilized. A total of 551 patients (61% male, mean age 55.5 ± 13.3 years) patients were divided into training, testing, and validation datasets of 441, 55 and 55 patients, respectively. The main candidate genes found were: FN1, ICAM1, MYD88, IL10, and CCL2 with the ANN model; and MMP9, MYD88, and CDK4 with LR model. The AUCs were 0.71 for the LR and 0.81 for the ANN analysis. Compared to the LR model, the ANN model showed better results: Accuracy rate, 83.3 %; H-L statistic, 6.5 %; and AUC, 0.81 % of patients. The findings show that ANNs can accurately predict the 15-month survival in GBM patients and contribute to precise medical treatment.

4.
BMC Infect Dis ; 24(1): 803, 2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39123113

RESUMO

BACKGROUND: Predicting an individual's risk of death from COVID-19 is essential for planning and optimising resources. However, since the real-world mortality rate is relatively low, particularly in places like Hong Kong, this makes building an accurate prediction model difficult due to the imbalanced nature of the dataset. This study introduces an innovative application of graph convolutional networks (GCNs) to predict COVID-19 patient survival using a highly imbalanced dataset. Unlike traditional models, GCNs leverage structural relationships within the data, enhancing predictive accuracy and robustness. By integrating demographic and laboratory data into a GCN framework, our approach addresses class imbalance and demonstrates significant improvements in prediction accuracy. METHODS: The cohort included all consecutive positive COVID-19 patients fulfilling study criteria admitted to 42 public hospitals in Hong Kong between January 23 and December 31, 2020 (n = 7,606). We proposed the population-based graph convolutional neural network (GCN) model which took blood test results, age and sex as inputs to predict the survival outcomes. Furthermore, we compared our proposed model to the Cox Proportional Hazard (CPH) model, conventional machine learning models, and oversampling machine learning models. Additionally, a subgroup analysis was performed on the test set in order to acquire a deeper understanding of the relationship between each patient node and its neighbours, revealing possible underlying causes of the inaccurate predictions. RESULTS: The GCN model was the top-performing model, with an AUC of 0.944, considerably outperforming all other models (p < 0.05), including the oversampled CPH model (0.708), linear regression (0.877), Linear Discriminant Analysis (0.860), K-nearest neighbours (0.834), Gaussian predictor (0.745) and support vector machine (0.847). With Kaplan-Meier estimates, the GCN model demonstrated good discriminability between low- and high-risk individuals (p < 0.0001). Based on subanalysis using the weighted-in score, although the GCN model was able to discriminate well between different predicted groups, the separation was inadequate between false negative (FN) and true negative (TN) groups. CONCLUSION: The GCN model considerably outperformed all other machine learning methods and baseline CPH models. Thus, when applied to this imbalanced COVID survival dataset, adopting a population graph representation may be an approach to achieving good prediction.


Assuntos
COVID-19 , Redes Neurais de Computação , SARS-CoV-2 , Humanos , COVID-19/mortalidade , COVID-19/diagnóstico , Masculino , Feminino , Pessoa de Meia-Idade , Hong Kong/epidemiologia , Idoso , Adulto , Testes Hematológicos/métodos , Aprendizado de Máquina , Modelos de Riscos Proporcionais , Estudos de Coortes
5.
Quant Imaging Med Surg ; 14(8): 5408-5419, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39144008

RESUMO

Background: Automated tumor segmentation and survival prediction are critical to clinical diagnosis and treatment. This study aimed to develop deep-learning models for automatic tumor segmentation and survival prediction in magnetic resonance imaging (MRI) of cervical cancer (CC) by combining deep neural networks and Transformer architecture. Methods: This study included 406 patients with CC, each with comprehensive clinical information and MRI scans. We randomly divided patients into training, validation, and independent test cohorts in a 6:2:2 ratio. During the model training, we employed two architecture types: one being a hybrid model combining convolutional neural network (CNN) and ransformer (CoTr) and one of pure CNNs. For survival prediction, the hybrid model combined tumor image features extracted by segmentation models with clinical information. The performance of the segmentation models was evaluated using the Dice similarity coefficient (DSC) and 95% Hausdorff distance (HD95). The performance of the survival models was assessed using the concordance index. Results: The CoTr model performed well in both contrast-enhanced T1-weighted (ceT1W) and T2-weighted (T2W) imaging segmentation tasks, with average DSCs of 0.827 and 0.820, respectively, which outperformed other the CNN models such as U-Net (DSC: 0.807 and 0.808), attention U-Net (DSC: 0.814 and 0.811), and V-Net (DSC: 0.805 and 0.807). For survival prediction, the proposed deep-learning model significantly outperformed traditional methods, yielding a concordance index of 0.732. Moreover, it effectively divided patients into low-risk and high-risk groups for disease progression (P<0.001). Conclusions: Combining Transformer architecture with a CNN can improve MRI tumor segmentation, and this deep-learning model excelled in the survival prediction of patients with CC as compared to traditional methods.

6.
Immunooncol Technol ; 24: 100723, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39185322

RESUMO

Background: Integrating complementary diagnostic data sources promises enhanced robustness in the predictive performance of artificial intelligence (AI) models, a crucial requirement for future clinical validation/implementation. In this study, we investigate the potential value of integrating data from noninvasive diagnostic modalities, including chest computed tomography (CT) imaging, routine laboratory blood tests, and clinical parameters, to retrospectively predict 1-year survival in a cohort of patients with advanced non-small-cell lung cancer, melanoma, and urothelial cancer treated with immunotherapy. Patients and methods: The study included 475 patients, of whom 444 had longitudinal CT scans and 475 had longitudinal laboratory data. An ensemble of AI models was trained on data from each diagnostic modality, and subsequently, a model-agnostic integration approach was adopted for combining the prediction probabilities of each modality and producing an integrated decision. Results: Integrating different diagnostic data demonstrated a modest increase in predictive performance. The highest area under the curve (AUC) was achieved by CT and laboratory data integration (AUC of 0.83, 95% confidence interval 0.81-0.85, P < 0.001), whereas the performance of individual models trained on laboratory and CT data independently yielded AUCs of 0.81 and 0.73, respectively. Conclusions: In our retrospective cohort, integrating different noninvasive data modalities improved performance.

7.
Front Genet ; 15: 1378809, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39161422

RESUMO

Introduction: Developing effective breast cancer survival prediction models is critical to breast cancer prognosis. With the widespread use of next-generation sequencing technologies, numerous studies have focused on survival prediction. However, previous methods predominantly relied on single-omics data, and survival prediction using multi-omics data remains a significant challenge. Methods: In this study, considering the similarity of patients and the relevance of multi-omics data, we propose a novel multi-omics stacked fusion network (MSFN) based on a stacking strategy to predict the survival of breast cancer patients. MSFN first constructs a patient similarity network (PSN) and employs a residual graph neural network (ResGCN) to obtain correlative prognostic information from PSN. Simultaneously, it employs convolutional neural networks (CNNs) to obtain specificity prognostic information from multi-omics data. Finally, MSFN stacks the prognostic information from these networks and feeds into AdaboostRF for survival prediction. Results: Experiments results demonstrated that our method outperformed several state-of-the-art methods, and biologically validated by Kaplan-Meier and t-SNE.

9.
Front Oncol ; 14: 1396726, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39055563

RESUMO

Background: Prognostic assessment for colorectal cancer (CRC) displays substantial heterogeneity, as reliance solely on traditional TNM staging falls short of achieving precise individualized predictions. The integration of diverse biological information sources holds the potential to enhance prognostic accuracy. Objective: To establish a comprehensive multi-tiered precision prognostic evaluation system for CRC by amalgamating gene expression profiles, clinical characteristics, and tumor microsatellite instability (MSI) status in CRC patients. Methods: We integrated genomic data, clinical information, and survival follow-up data from 483 CRC patients obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. MSI-related gene modules were identified using differential expression analysis and Weighted Gene Co-expression Network Analysis (WGCNA). Three prognostic models were constructed: MSI-Related Gene Prognostic Model (Model I), Clinical Prognostic Model (Model II), and Integrated Multi-Layered Prognostic Model (Model III) by combining clinical features. Model performance was assessed and compared using Receiver Operating Characteristic (ROC) curves, Kaplan-Meier analysis, and other methods. Results: Six MSI-related genes were selected for constructing Model I (AUC = 0.724); Model II used two clinical features (AUC = 0.684). Compared to individual models, the integrated Model III exhibited superior performance (AUC = 0.825) and demonstrated good stability in an independent dataset (AUC = 0.767). Conclusion: This study successfully developed and validated a comprehensive multi-tiered precision prognostic assessment model for CRC, providing an effective tool for personalized medical management of CRC.

10.
Sci Rep ; 14(1): 17064, 2024 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-39048590

RESUMO

Deep learning (DL) has shown potential to provide powerful representations of bulk RNA-seq data in cancer research. However, there is no consensus regarding the impact of design choices of DL approaches on the performance of the learned representation, including the model architecture, the training methodology and the various hyperparameters. To address this problem, we evaluate the performance of various design choices of DL representation learning methods using TCGA and DepMap pan-cancer datasets and assess their predictive power for survival and gene essentiality predictions. We demonstrate that baseline methods achieve comparable or superior performance compared to more complex models on survival predictions tasks. DL representation methods, however, are the most efficient to predict the gene essentiality of cell lines. We show that auto-encoders (AE) are consistently improved by techniques such as masking and multi-head training. Our results suggest that the impact of DL representations and of pretraining are highly task- and architecture-dependent, highlighting the need for adopting rigorous evaluation guidelines. These guidelines for robust evaluation are implemented in a pipeline made available to the research community.


Assuntos
Aprendizado Profundo , Genes Essenciais , RNA-Seq , Humanos , RNA-Seq/métodos , Neoplasias/genética , Neoplasias/mortalidade , Biologia Computacional/métodos
11.
Artigo em Inglês | MEDLINE | ID: mdl-38974963

RESUMO

Severe cases of COVID-19 often necessitate escalation to the Intensive Care Unit (ICU), where patients may face grave outcomes, including mortality. Chest X-rays play a crucial role in the diagnostic process for evaluating COVID-19 patients. Our collaborative efforts with Michigan Medicine in monitoring patient outcomes within the ICU have motivated us to investigate the potential advantages of incorporating clinical information and chest X-ray images for predicting patient outcomes. We propose an analytical workflow to address challenges such as the absence of standardized approaches for image pre-processing and data utilization. We then propose an ensemble learning approach designed to maximize the information derived from multiple prediction algorithms. This entails optimizing the weights within the ensemble and considering the common variability present in individual risk scores. Our simulations demonstrate the superior performance of this weighted ensemble averaging approach across various scenarios. We apply this refined ensemble methodology to analyze post-ICU COVID-19 mortality, an occurrence observed in 21% of COVID-19 patients admitted to the ICU at Michigan Medicine. Our findings reveal substantial performance improvement when incorporating imaging data compared to models trained solely on clinical risk factors. Furthermore, the addition of radiomic features yields even larger enhancements, particularly among older and more medically compromised patients. These results may carry implications for enhancing patient outcomes in similar clinical contexts.

12.
Comput Methods Programs Biomed ; 255: 108338, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39042996

RESUMO

BACKGROUND AND OBJECTIVE: Patients with glioblastoma have a five-year relative survival rate of less than 5 %. Thus, accurately predicting the overall survival (OS) of patients with glioblastoma is crucial for effective treatment planning. METHODS: To fully leverage the imaging characteristics of glioblastomas, we propose a segmentation-guided regression method for predicting OS of patients with brain tumors using multimodal magnetic resonance imaging. Specifically, a brain tumor segmentation network was first pre-trained without leveraging survival information. Subsequently, the survival regression network was jointly trained with the guidance of brain tumor segmentation, focusing on tumor voxels and suppressing irrelevant backgrounds. RESULTS: Our proposed framework, based on the well-known backbone of UNETR++, achieved a Dice score of 0.7910, Spearman correlation of 0.4112, and Harrell's concordance index of 0.6488. The model consistently showed promising results compared with baseline methods on two different datasets (BraTS and UCSF-PDGM). Furthermore, ablation studies on our training configurations demonstrated that both the pre-training segmentation network and contrastive loss significantly improved all metrics for OS prediction. CONCLUSIONS: In this study, we propose a joint learning framework based on a pre-trained segmentation backbone for OS prediction by leveraging a brain tumor segmentation map. By utilizing a spatial feature map, our model can operate using a sliding-window approach, which can be adopted by varying the matrix sizes and resolutions of the input images.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Imageamento por Ressonância Magnética , Glioblastoma/diagnóstico por imagem , Glioblastoma/mortalidade , Humanos , Imageamento por Ressonância Magnética/métodos , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/mortalidade , Imagem Multimodal/métodos , Algoritmos , Processamento de Imagem Assistida por Computador/métodos , Prognóstico , Redes Neurais de Computação
13.
Transl Cancer Res ; 13(6): 2751-2766, 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38988930

RESUMO

Background: Pancreatic ductal adenocarcinoma (PDAC), which accounts for the vast majority of pancreatic cancer (PC), is a highly aggressive malignancy with a dismal prognosis. Age is shown to be an independent factor affecting survival outcomes in patients with PDAC. Our study aimed to identify prognostic factors and construct a nomogram to predict survival in PDAC patients aged ≥60 years. Methods: Data of PDAC patients aged ≥60 years were collected from the Surveillance, Epidemiology, and End Results (SEER) database. Multivariate Cox regression analysis was used to determined prognostic factors of overall survival (OS) and cancer-specific survival (CSS), and two nomograms were constructed and validated by calibration plots, concordance index (C-index) and decision curve analysis (DCA). Additionally, 432 patients from the First Affiliated Hospital of Wenzhou Medical University were included as an external cohort. Kaplan-Meier curves were applied to further verify the clinical validity of the nomograms. Results: Ten independent prognostic factors were identified to establish the nomograms. The C-indexes of the training and validation groups based on the OS nomogram were 0.759 and 0.760, higher than those of the tumor-node-metastasis (TNM) staging system (0.638 and 0.636, respectively). Calibration curves showed high consistency between predictions and observations. Better area under the receiver operator characteristic (ROC) curve (AUC) values and DCA were also obtained compared to the TNM system. The risk stratification based on the nomogram could distinguish patients with different survival risks. Conclusions: We constructed and externally validated a population-based survival-predicting nomogram for PDAC patients aged ≥60 years. The new model could help clinicians personalize survival prediction and risk assessment.

14.
Front Artif Intell ; 7: 1428501, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39021434

RESUMO

Survival prediction integrates patient-specific molecular information and clinical signatures to forecast the anticipated time of an event, such as recurrence, death, or disease progression. Survival prediction proves valuable in guiding treatment decisions, optimizing resource allocation, and interventions of precision medicine. The wide range of diseases, the existence of various variants within the same disease, and the reliance on available data necessitate disease-specific computational survival predictors. The widespread adoption of artificial intelligence (AI) methods in crafting survival predictors has undoubtedly revolutionized this field. However, the ever-increasing demand for more sophisticated and effective prediction models necessitates the continued creation of innovative advancements. To catalyze these advancements, it is crucial to bring existing survival predictors knowledge and insights into a centralized platform. The paper in hand thoroughly examines 23 existing review studies and provides a concise overview of their scope and limitations. Focusing on a comprehensive set of 90 most recent survival predictors across 44 diverse diseases, it delves into insights of diverse types of methods that are used in the development of disease-specific predictors. This exhaustive analysis encompasses the utilized data modalities along with a detailed analysis of subsets of clinical features, feature engineering methods, and the specific statistical, machine or deep learning approaches that have been employed. It also provides insights about survival prediction data sources, open-source predictors, and survival prediction frameworks.

15.
Sci Rep ; 14(1): 15004, 2024 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-38951567

RESUMO

The tumor microenvironment (TME) plays a fundamental role in tumorigenesis, tumor progression, and anti-cancer immunity potential of emerging cancer therapeutics. Understanding inter-patient TME heterogeneity, however, remains a challenge to efficient drug development. This article applies recent advances in machine learning (ML) for survival analysis to a retrospective study of NSCLC patients who received definitive surgical resection and immune pathology following surgery. ML methods are compared for their effectiveness in identifying prognostic subtypes. Six survival models, including Cox regression and five survival machine learning methods, were calibrated and applied to predict survival for NSCLC patients based on PD-L1 expression, CD3 expression, and ten baseline patient characteristics. Prognostic subregions of the biomarker space are delineated for each method using synthetic patient data augmentation and compared between models for overall survival concordance. A total of 423 NSCLC patients (46% female; median age [inter quantile range]: 67 [60-73]) treated with definite surgical resection were included in the study. And 219 (52%) patients experienced events during the observation period consisting of a maximum follow-up of 10 years and median follow up 78 months. The random survival forest (RSF) achieved the highest predictive accuracy, with a C-index of 0.84. The resultant biomarker subtypes demonstrate that patients with high PD-L1 expression combined with low CD3 counts experience higher risk of death within five-years of surgical resection.


Assuntos
Biomarcadores Tumorais , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Aprendizado de Máquina , Microambiente Tumoral , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Feminino , Masculino , Idoso , Pessoa de Meia-Idade , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/cirurgia , Prognóstico , Estudos Retrospectivos , Biomarcadores Tumorais/metabolismo , Antígeno B7-H1/metabolismo , Análise de Sobrevida
16.
Artigo em Inglês | MEDLINE | ID: mdl-38967327

RESUMO

This study attempted to build a prognostic riskscore model for pancreatic cancer (PC) patients based on vesicle-mediated transport protein-related genes (VMTGs). We initially conducted differential expression analysis and Cox regression analysis, followed by the construction of a riskscore model to classify PC patients into high-risk (HR) and low-risk (LR) groups. The GEO GSE62452 dataset further validated the model. Kaplan-Meier survival analysis was employed to analyze the survival rate of the HR group and LR group. Cox analysis confirmed the independent prognostic ability of the riskscore model. Additionally, we evaluated immune status in both HR and LR groups, utilizing data from the GDSC database to predict drug response among PC patients. We identified six PC-specific genes from 724 VMTGs. Survival analysis revealed that the survival rate of the HR group was lower than that of the LR group (P<0.05). Cox analysis confirmed that the prognostic riskscore model could independently predict the survival status of PC patients (P<0.001). Immunological analysis revealed that the ESTIMATE score, immune score, and stroma score of the HR group were considerably lower than those of the LR group, and the tumor purity score of the HR group was higher. The IC50 values of Gemcitabine, Irinotecan, Oxaliplatin, and Paclitaxel in the LR group were considerably lower than those in the HR group (P<0.001). In summary, the VMTG-based prognostic riskscore model could stratify PC risk and effectively predict the survival of PC patients.

17.
Cancers (Basel) ; 16(13)2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-39001463

RESUMO

Survival prediction post-cystectomy is essential for the follow-up care of bladder cancer patients. This study aimed to evaluate artificial intelligence (AI)-large language models (LLMs) for extracting clinical information and improving image analysis, with an initial application involving predicting five-year survival rates of patients after radical cystectomy for bladder cancer. Data were retrospectively collected from medical records and CT urograms (CTUs) of bladder cancer patients between 2001 and 2020. Of 781 patients, 163 underwent chemotherapy, had pre- and post-chemotherapy CTUs, underwent radical cystectomy, and had an available post-surgery five-year survival follow-up. Five AI-LLMs (Dolly-v2, Vicuna-13b, Llama-2.0-13b, GPT-3.5, and GPT-4.0) were used to extract clinical descriptors from each patient's medical records. As a reference standard, clinical descriptors were also extracted manually. Radiomics and deep learning descriptors were extracted from CTU images. The developed multi-modal predictive model, CRD, was based on the clinical (C), radiomics (R), and deep learning (D) descriptors. The LLM retrieval accuracy was assessed. The performances of the survival predictive models were evaluated using AUC and Kaplan-Meier analysis. For the 163 patients (mean age 64 ± 9 years; M:F 131:32), the LLMs achieved extraction accuracies of 74%~87% (Dolly), 76%~83% (Vicuna), 82%~93% (Llama), 85%~91% (GPT-3.5), and 94%~97% (GPT-4.0). For a test dataset of 64 patients, the CRD model achieved AUCs of 0.89 ± 0.04 (manually extracted information), 0.87 ± 0.05 (Dolly), 0.83 ± 0.06~0.84 ± 0.05 (Vicuna), 0.81 ± 0.06~0.86 ± 0.05 (Llama), 0.85 ± 0.05~0.88 ± 0.05 (GPT-3.5), and 0.87 ± 0.05~0.88 ± 0.05 (GPT-4.0). This study demonstrates the use of LLM model-extracted clinical information, in conjunction with imaging analysis, to improve the prediction of clinical outcomes, with bladder cancer as an initial example.

18.
Artigo em Inglês | MEDLINE | ID: mdl-38969836

RESUMO

Heart failure (HF) is associated with high rates of morbidity and mortality. The value of deep learning survival prediction models using chest radiographs in patients with heart failure is currently unclear. The aim of our study is to develop and validate a deep learning survival prediction model using chest X-ray (DLSPCXR) in patients with HF. The study retrospectively enrolled a cohort of 353 patients with HF who underwent chest X-ray (CXR) at our institution between March 2012 and March 2017. The dataset was randomly divided into training (n = 247) and validation (n = 106) datasets. Univariate and multivariate Cox analysis were conducted on the training dataset to develop clinical and imaging survival prediction models. The DLSPCXR was trained and the selected clinical parameters were incorporated into DLSPCXR to establish a new model called DLSPinteg. Discrimination performance was evaluated using the time-dependent area under the receiver operating characteristic curves (TD AUC) at 1, 3, and 5-years survival. Delong's test was employed for the comparison of differences between two AUCs of different models. The risk-discrimination capability of the optimal model was evaluated by the Kaplan-Meier curve. In multivariable Cox analysis, older age, higher N-terminal pro-B-type natriuretic peptide (NT-ProBNP), systolic pulmonary artery pressure (sPAP) > 50 mmHg, New York Heart Association (NYHA) functional class III-IV and cardiothoracic ratio (CTR) ≥ 0.62 in CXR were independent predictors of poor prognosis in patients with HF. Based on the receiver operating characteristic (ROC) curve analysis, DLSPCXR had better performance at predicting 5-year survival than the imaging Cox model in the validation cohort (AUC: 0.757 vs. 0.561, P = 0.01). DLSPinteg as the optimal model outperforms the clinical Cox model (AUC: 0.826 vs. 0.633, P = 0.03), imaging Cox model (AUC: 0.826 vs. 0.555, P < 0.001), and DLSPCXR (AUC: 0.826 vs. 0.767, P = 0.06). Deep learning models using chest radiographs can predict survival in patients with heart failure with acceptable accuracy.

19.
Heliyon ; 10(11): e31873, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38845954

RESUMO

Background: Survival prediction is one of the crucial goals in precision medicine, as accurate survival assessment can aid physicians in selecting appropriate treatment for individual patients. To achieve this aim, extensive data must be utilized to train the prediction model and prevent overfitting. However, the collection of patient data for disease prediction is challenging due to potential variations in data sources across institutions and concerns regarding privacy and ownership issues in data sharing. To facilitate the integration of cancer data from different institutions without violating privacy laws, we developed a federated learning-based data integration framework called AdFed, which can be used to evaluate patients' survival while considering the privacy protection problem by utilizing the decentralized federated learning technology and regularization method. Results: AdFed was tested on different cancer datasets that contain the patients' information from different institutions. The experimental results show that AdFed using distributed data can achieve better performance in cancer survival prediction (AUC = 0.605) than the compared federated-learning-based methods (average AUC = 0.554). Additionally, to assess the biological interpretability of our method, in the case study we list 10 identified genes related to liver cancer selected by AdFed, among which 5 genes have been proved by literature review. Conclusions: The results indicate that AdFed outperforms better than other federated-learning-based methods, and the interpretable algorithm can select biologically significant genes and pathways while ensuring the confidentiality and integrity of data.

20.
Sci Rep ; 14(1): 12934, 2024 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-38839983

RESUMO

Osteosarcoma is a primary malignant tumor that commonly affects children and adolescents, with a poor prognosis. The existence of tumor heterogeneity leads to different molecular subtypes and survival outcomes. Recently, lipid metabolism has been identified as a critical characteristic of cancer. Therefore, our study aims to identify osteosarcoma's lipid metabolism molecular subtype and develop a signature for survival outcome prediction. Four multicenter cohorts-TARGET-OS, GSE21257, GSE39058, and GSE16091-were amalgamated into a unified Meta-Cohort. Through consensus clustering, novel molecular subtypes within Meta-Cohort patients were delineated. Subsequent feature selection processes, encompassing analyses of differentially expressed genes between subtypes, univariate Cox analysis, and StepAIC, were employed to pinpoint biomarkers related to lipid metabolism in TARGET-OS. We selected the most effective algorithm for constructing a Lipid Metabolism-Related Signature (LMRS) by utilizing four machine-learning algorithms reconfigured into ten unique combinations. This selection was based on achieving the highest concordance index (C-index) in the test cohort of GSE21257, GSE39058, and GSE16091. We identified two distinct lipid metabolism molecular subtypes in osteosarcoma patients, C1 and C2, with significantly different survival rates. C1 is characterized by increased cholesterol, fatty acid synthesis, and ketone metabolism. In contrast, C2 focuses on steroid hormone biosynthesis, arachidonic acid, and glycerolipid and linoleic acid metabolism. Feature selection in the TARGET-OS identified 12 lipid metabolism genes, leading to a model predicting osteosarcoma patient survival. The LMRS, based on the 12 identified genes, consistently accurately predicted prognosis across TARGET-OS, testing cohorts, and Meta-Cohort. Incorporating 12 published signatures, LMRS showed robust and significantly superior predictive capability. Our results offer a promising tool to enhance the clinical management of osteosarcoma, potentially leading to improved clinical outcomes.


Assuntos
Neoplasias Ósseas , Metabolismo dos Lipídeos , Aprendizado de Máquina , Osteossarcoma , Osteossarcoma/genética , Osteossarcoma/mortalidade , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Humanos , Metabolismo dos Lipídeos/genética , Prognóstico , Neoplasias Ósseas/genética , Neoplasias Ósseas/mortalidade , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Feminino , Masculino , Regulação Neoplásica da Expressão Gênica , Adolescente , Perfilação da Expressão Gênica/métodos , Criança
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA