Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Phys Med Biol ; 69(7)2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38224617

ABSTRACT

Objective.In the realm of utilizing artificial intelligence (AI) for medical image analysis, the paradigm of 'signal-image-knowledge' has remained unchanged. However, the process of 'signal to image' inevitably introduces information distortion, ultimately leading to irrecoverable biases in the 'image to knowledge' process. Our goal is to skip reconstruction and build a diagnostic model directly from the raw data (signal).Approach. This study focuses on computed tomography (CT) and its raw data (sinogram) as the research subjects. We simulate the real-world process of 'human-signal-image' using the workflow 'CT-simulated data- reconstructed CT,' and we develop a novel AI predictive model directly targeting raw data (RCTM). This model comprises orientation, spatial, and global analysis modules, embodying the fusion of local to global information extraction from raw data. We selected 1994 patients with retrospective cases of solid lung nodules and modeled different types of data.Main results. We employed predefined radiomic features to assess the diagnostic feature differences caused by reconstruction. The results indicated that approximately 14% of the features had Spearman correlation coefficients below 0.8. These findings suggest that despite the increasing maturity of CT reconstruction algorithms, they still introduce perturbations to diagnostic features. Moreover, our proposed RCTM achieved an area under the curve (AUC) of 0.863 in the diagnosis task, showcasing a comprehensive superiority over models constructed from secondary reconstructed CTs (0.840, 0.822, and 0.825). Additionally, the performance of RCTM closely resembled that of models constructed from original CT scans (0.868, 0.878, and 0.866).Significance. The diagnostic and therapeutic approach directly based on CT raw data can enhance the precision of AI models and the concept of 'signal-to-image' can be extended to other types of imaging. AI diagnostic models tailored to raw data offer the potential to disrupt the traditional paradigm of 'signal-image-knowledge', opening up new avenues for more accurate medical diagnostics.


Subject(s)
Artificial Intelligence , Radiology , Humans , Retrospective Studies , Tomography, X-Ray Computed/methods , Algorithms , Image Processing, Computer-Assisted/methods
2.
Eur J Med Res ; 28(1): 545, 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-38017548

ABSTRACT

BACKGROUND: umor cells, immune cells and stromal cells jointly modify tumor development and progression. We aim to explore the potential effects of tumor purity on the immune microenvironment, genetic landscape and prognosis in prostate cancer (PCa). METHODS: Tumor purity of prostate cancer patients was extracted from The cancer genome atlas (TCGA). Immune cellular proportions were calculated by the CIBERSORT. To identify critical modules related to tumor purity, we used weighted gene co-expression network analysis (WGCNA). Using STRING and Cytoscape, protein-protein interaction (PPI) networks were constructed and analyzed. A Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, Disease Ontology (DO), and Gene Set Enrichment Analysis (GSEA) enrichment analysis of identified modules was conducted. To identify the expression of key genes at protein levels, we used the Human Protein Atlas (HPA) platform. RESULTS: A model of tumor purity score (TPS) was constructed in the gene expression omnibus series (GSE) 116,918 cohort. TCGA cohort served as a validation set and was employed to validate the TPS. TPS model, as an independent prognostic factor of distant metastasis-free survival (DMFS) in PCa. Patients had higher tumor purity and better prognosis in the low-TPS group. Tumor purity was related to the infiltration of mast cells and macrophage cells positively, whereas related to the infiltration of dendritic cells, T cells and B cells negatively in PCa. The nomogram based on TPS, Age, Gleason score and T stage had a good predictive value and could evaluate the prognosis of PCa metastasis. GO and KEGG enrichment analyses showed that hub genes mainly participate in T cell activation and T-helper lymphocytes (TH) differentiation. Hub genes were mainly enriched in primary immunodeficiency disease, according to DO analysis. SLAMF8 was identified as the most critical gene by Cytoscape and HPA analysis. CONCLUSIONS: Dynamic changes in the immune microenvironment associated with tumor purity could correlate with a poor DMFS of low-purity PCa. The TPS can predict the DMFS of PCa. In addition, prostate cancer metastases may be related to immunosuppression caused by a disorder of the immune microenvironment.


Subject(s)
Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/genetics , Cell Differentiation , Gene Expression Profiling , Gene Ontology , Lymphocyte Activation , Tumor Microenvironment/genetics , Signaling Lymphocytic Activation Molecule Family
3.
Neural Netw ; 164: 455-463, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37182347

ABSTRACT

Prognostic prediction has long been a hotspot in disease analysis and management, and the development of image-based prognostic prediction models has significant clinical implications for current personalized treatment strategies. The main challenge in prognostic prediction is to model a regression problem based on censored observations, and semi-supervised learning has the potential to play an important role in improving the utilization efficiency of censored data. However, there are yet few effective semi-supervised paradigms to be applied. In this paper, we propose a semi-supervised co-training deep neural network incorporating a support vector regression layer for survival time estimation (Co-DeepSVS) that improves the efficiency in utilizing censored data for prognostic prediction. First, we introduce a support vector regression layer in deep neural networks to deal with censored data and directly predict survival time, and more importantly to calculate the labeling confidence of each case. Then, we apply a semi-supervised multi-view co-training framework to achieve accurate prognostic prediction, where labeling confidence estimation with prior knowledge of pseudo time is conducted for each view. Experimental results demonstrate that the proposed Co-DeepSVS has a promising prognostic ability and surpasses most widely used methods on a multi-phase CT dataset. Besides, the introduction of SVR layer makes the model more robust in the presence of follow-up bias.


Subject(s)
Knowledge , Neural Networks, Computer , Prognosis , Supervised Machine Learning
4.
Comput Biol Med ; 158: 106809, 2023 05.
Article in English | MEDLINE | ID: mdl-37004433

ABSTRACT

Projection magnetic particle imaging (MPI) can significantly improve the temporal resolution of three-dimensional (3D) imaging compared to that using traditional point by point scanning. However, the dense view of projections required for tomographic reconstruction limits the scope of temporal resolution optimization. The solution to this problem in computed tomography (CT) is using limited view projections (sparse view or limited angle) for reconstruction, which can be divided into: completing the limited view sinogram and image post-processing for streaking artifacts caused by insufficient projections. Benefiting from large-scale CT datasets, both categories of deep learning-based methods have achieved tremendous progress; yet, there is a data scarcity limitation in MPI. We propose a cross-domain knowledge transfer learning strategy that can transfer the prior knowledge of the limited view learned by the model in CT to MPI, which can help reduce the network requirements for real MPI data. In addition, the size of the imaging target affects the scale of the streaking artifacts caused by insufficient projections. Therefore, we propose a parallel-cascaded multi-scale attention module that allows the network to adaptively identify streaking artifacts at different scales. The proposed method was evaluated on real phantom and in vivo mouse data, and it significantly outperformed several advanced limited view methods. The streaking artifacts caused by an insufficient number of projections can be overcome using the proposed method.


Subject(s)
Algorithms , Artifacts , Animals , Mice , Tomography, X-Ray Computed/methods , Phantoms, Imaging , Magnetic Phenomena , Image Processing, Computer-Assisted/methods
5.
Med Phys ; 50(4): 2354-2371, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36239207

ABSTRACT

BACKGROUND: Magnetic particle imaging (MPI) is a novel tomographic imaging modality that scans the distribution of superparamagnetic iron oxide nanoparticles. However, it is time-consuming to scan multiview two-dimensional (2D) projections for three-dimensional (3D) reconstruction in projection MPI, such as computed tomography (CT). An intuitive idea is to use the sparse-view projections for reconstruction to improve the temporal resolution. Tremendous progress has been made toward addressing the sparse-view problem in CT, because of the availability of large data sets. For the novel tomography of MPI, to the best of our knowledge, studies on the sparse-view problem have not yet been reported. PURPOSE: The acquisition of multiview projections for 3D MPI imaging is time-consuming. Our goal is to only acquire sparse-view projections for reconstruction to improve the 3D imaging temporal resolution of projection MPI. METHODS: We propose to address the sparse-view problem in projection MPI by generating novel projections. The data set we constructed consists of three parts: simulation data set (including 3000 3D data), four phantoms data, and an in vivo mouse data. The simulation data set is used to train and validate the network, and the phantoms and in vivo mouse data are used to test the network. When the number of novel generated projections meets the requirements of filtered back projection, the streaking artifacts will be absent from MPI tomographic imaging. Specifically, we propose a projection generative network (PGNet), that combines an attention mechanism, adversarial training strategy, and a fusion loss function and can generate novel projections based on sparse-view real projections. To the best of our knowledge, we are the first to propose a deep learning method to attempt to overcome the sparse-view problem in projection MPI. RESULTS: We compare our method with several sparse-view methods on phantoms and in vivo mouse data and validate the advantages and effectiveness of our proposed PGNet. Our proposed PGNet enables the 3D imaging temporal resolution of projection MPI to be improved by 6.6 times, while significantly suppressing the streaking artifacts. CONCLUSION: We proposed a deep learning method operated in projection domain to address the sparse-view reconstruction of MPI, and the data scarcity problem in projection MPI reconstruction is alleviated by constructing a sparse-dense simulated projection data set. By our proposed method, the number of acquisitions of real projections can be reduced. The advantage of our method is that it prevents the generation of streaking artifacts at the source. Our proposed sparse-view reconstruction method has great potential for application to time-sensitive in vivo 3D MPI imaging.


Subject(s)
Tomography, X-Ray Computed , Tomography , Animals , Mice , Tomography, X-Ray Computed/methods , Imaging, Three-Dimensional/methods , Phantoms, Imaging , Magnetic Phenomena , Image Processing, Computer-Assisted/methods , Algorithms
6.
Health Data Sci ; 3: 0005, 2023.
Article in English | MEDLINE | ID: mdl-38487199

ABSTRACT

Importance: Digestive system neoplasms (DSNs) are the leading cause of cancer-related mortality with a 5-year survival rate of less than 20%. Subjective evaluation of medical images including endoscopic images, whole slide images, computed tomography images, and magnetic resonance images plays a vital role in the clinical practice of DSNs, but with limited performance and increased workload of radiologists or pathologists. The application of artificial intelligence (AI) in medical image analysis holds promise to augment the visual interpretation of medical images, which could not only automate the complicated evaluation process but also convert medical images into quantitative imaging features that associated with tumor heterogeneity. Highlights: We briefly introduce the methodology of AI for medical image analysis and then review its clinical applications including clinical auxiliary diagnosis, assessment of treatment response, and prognosis prediction on 4 typical DSNs including esophageal cancer, gastric cancer, colorectal cancer, and hepatocellular carcinoma. Conclusion: AI technology has great potential in supporting the clinical diagnosis and treatment decision-making of DSNs. Several technical issues should be overcome before its application into clinical practice of DSNs.

7.
EBioMedicine ; 86: 104364, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36395737

ABSTRACT

BACKGROUND: This study, based on multicentre cohorts, aims to utilize computed tomography (CT) images to construct a deep learning model for predicting major pathological response (MPR) to neoadjuvant chemoimmunotherapy in non-small cell lung cancer (NSCLC) and further explore the biological basis under its prediction. METHODS: 274 patients undergoing curative surgery after neoadjuvant chemoimmunotherapy for NSCLC at 4 centres from January 2019 to December 2021 were included and divided into a training cohort, an internal validation cohort, and an external validation cohort. ShuffleNetV2x05-based features of the primary tumour on the CT scans within the 2 weeks preceding neoadjuvant administration were employed to develop a deep learning score for distinguishing MPR and non-MPR. To reveal the underlying biological basis of the deep learning score, a genetic analysis was conducted based on 25 patients with RNA-sequencing data. FINDINGS: MPR was achieved in 54.0% (n = 148) patients. The area under the curve (AUC) of the deep learning score to predict MPR was 0.73 (95% confidence interval [CI]: 0.58-0.86) and 0.72 (95% CI: 0.58-0.85) in the internal validation and external validation cohorts, respectively. After integrating the clinical characteristic into the deep learning score, the combined model achieved satisfactory performance in the internal validation (AUC: 0.77, 95% CI: 0.64-0.89) and external validation cohorts (AUC: 0.75, 95% CI: 0.62-0.87). In the biological basis exploration for the deep learning score, a high deep learning score was associated with the downregulation of pathways mediating tumour proliferation and the promotion of antitumour immune cell infiltration in the microenvironment. INTERPRETATION: The proposed deep learning model could effectively predict MPR in NSCLC patients treated with neoadjuvant chemoimmunotherapy. FUNDING: This study was supported by National Key Research and Development Program of China, China (2017YFA0205200); National Natural Science Foundation of China, China (91959126, 82022036, 91959130, 81971776, 81771924, 6202790004, 81930053, 9195910169, 62176013, 8210071009); Beijing Natural Science Foundation, China (L182061); Strategic Priority Research Program of Chinese Academy of Sciences, China (XDB38040200); Chinese Academy of Sciences, China (GJJSTD20170004, QYZDJ-SSW-JSC005); Shanghai Hospital Development Center, China (SHDC2020CR3047B); and Science and Technology Commission of Shanghai Municipality, China (21YF1438200).


Subject(s)
Carcinoma, Non-Small-Cell Lung , Deep Learning , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Neoadjuvant Therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Retrospective Studies , China , Tumor Microenvironment
8.
Transl Lung Cancer Res ; 11(4): 670-685, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35529789

ABSTRACT

Background: Radiomics based on computed tomography (CT) images is potential in promoting individualized treatment of non-small cell lung cancer (NSCLC), however, its role in immunotherapy needs further exploration. The aim of this study was to develop a CT-based radiomics score to predict the efficacy of immune checkpoint inhibitor (ICI) monotherapy in patients with advanced NSCLC. Methods: Two hundred and thirty-six ICI-treated patients were retrospectively included and divided into a training cohort (n=188) and testing cohort (n=48) at a ratio of 8 to 2. The efficacy outcomes of ICI were evaluated based on overall survival (OS) and progression-free survival (PFS). We designed a survival network and combined it with a Cox regression model to obtain patients' OS risk score (OSRS) and PFS risk score (PFSRS). Results: Based on OSRS and PFSRS, patients were divided into high- and low-risk groups in the training cohort and the test cohort with distinctly different [training cohort, log-rank P<0.001, hazard ratio (HR): 4.14; test cohort, log-rank P=0.014, HR: 4.54] and PFS (training cohort, log-rank P<0.001, HR: 4.52; test cohort, log-rank P<0.001, HR: 6.64). Further joint evaluation of OSRS and PFSRS showed that both were significant in the Cox regression model (P<0.001), and multi-overall survival risk score (MOSRS) displayed more outstanding stratification capabilities than OSRS in both the training (P<0.001) and test cohorts (P=0.002). None of the clinical characteristics were significant in the Cox regression model, and the score that predicted the best immune response was not as good as the risk score from follow-up information in the performance of prognostic stratification. Conclusions: We developed a CT imaging-based score with the potential to become an independent prognostic factor to screen patients who would benefit from ICI treatment, which suggested that CT radiomics could be applied for individualized immunotherapy of NSCLC. Our findings should be further validated by future larger multicenter study.

9.
Comput Biol Med ; 144: 105318, 2022 05.
Article in English | MEDLINE | ID: mdl-35245698

ABSTRACT

BACKGROUND: Gleason score (GS) is one of the most critical predictors of diagnosing prostate cancer (PCa). The prostate gland, including both lesions and their microenvironment, may contain more comprehensive information about the PCa. We aimed to investigate the potential of prostate gland radiomic features in identifying Gleason scores (GS) < 7, = 7, and >7. METHODS: We retrospectively examined preoperative magnetic resonance imaging (MRI) results, clinical data, and postoperative pathological findings from 489 PCa patients. The three-dimensional (3D) and two-dimensional (2D) radiomic features were extracted from the manually segmented 3D prostate gland and its maximum 2D layer on MRI, respectively. Significant features were selected, and sequence signatures were then developed via multi-class linear regression (MLR) accordingly. Subsequently, 2D and 3D radiomic models were constructed by applying MLR to the combination of the sequence signatures, respectively. The stability of the significant features was discussed by their average ranking in the other 30 random cohorts. Based on our distance matrix algorithm, we generated different regions of interest to simulate the manual segmentation biases and discuss the model's tolerance to them. RESULTS: Our 2D model reached a C-index of 0.728 and an average area under the receiver operating characteristic curve of 0.794 in the validation cohort. The corresponding key features were stable, with an average ranking of the top 8.352% in 30 random cohorts, and the model could tolerate a segmentation boundary deviation of 2 mm without significant performance degradation. CONCLUSION: 2D prostate-gland-MRI-based radiomic features showed stable potential in identifying GS.


Subject(s)
Prostate , Prostatic Neoplasms , Humans , Magnetic Resonance Imaging/methods , Male , Neoplasm Grading , Prostate/diagnostic imaging , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Retrospective Studies , Tumor Microenvironment
10.
Transl Oncol ; 14(8): 101113, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33975178

ABSTRACT

PURPOSE: Radiomic models have been demonstrated to have acceptable discrimination capability for detecting lymph node metastasis (LNM). We aimed to develop a computed tomography-based radiomic model and validate its usefulness in the prediction of normal-sized LNM at node level in cervical cancer. METHODS: A total of 273 LNs of 219 patients from 10 centers were evaluated in this study. We randomly divided the LNs from the 2 centers with the largest number of LNs into the training and internal validation cohorts, and the rest as the external validation cohort. Radiomic features were extracted from the arterial and venous phase images. We trained an artificial neural network (ANN) to develop two single-phase models. A radiomic model reflecting the features of two-phase images was also built for directly predicting LNM in cervical cancer. Moreover, four state-of-the-art methods were used for comparison. The performance of all models was assessed using the area under the receiver operating characteristic curve (AUC). RESULTS: Among the models we built, the models combining the features of two phases surpassed the single-phase models, and the models generated by ANN had better performance than the others. We found that the radiomic model achieved the highest AUCs of 0.912 and 0.859 in the training and internal validation cohorts, respectively. In the external validation cohort, the AUC of the radiomic model was 0.800. CONCLUSION: We constructed a radiomic model that exhibited great ability in the prediction of LNM. The application of the model could optimize clinical staging and decision-making.

11.
Transl Lung Cancer Res ; 10(2): 955-964, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33718035

ABSTRACT

BACKGROUND: Micropapillary/solid (MP/S) growth patterns of lung adenocarcinoma are vital for making clinical decisions regarding surgical intervention. This study aimed to predict the presence of a MP/S component in lung adenocarcinoma using radiomics analysis. METHODS: Between January 2011 and December 2013, patients undergoing curative invasive lung adenocarcinoma resection were included. Using the "PyRadiomics" package, we extracted 90 radiomics features from the preoperative computed tomography (CT) images. Subsequently, four prediction models were built by utilizing conventional machine learning approaches fitting into radiomics analysis: a generalized linear model (GLM), Naïve Bayes, support vector machine (SVM), and random forest classifiers. The models' accuracy was assessed using a receiver operating curve (ROC) analysis, and the models' stability was validated both internally and externally. RESULTS: A total of 268 patients were included as a primary cohort, and 36.6% (98/268) of them had lung adenocarcinoma with an MP/S component. Patients with an MP/S component had a higher rate of lymph node metastasis (18.4% versus 5.3%) and worse recurrence-free and overall survival. Five radiomics features were selected for model building, and in the internal validation, the four models achieved comparable performance of MP/S prediction in terms of area under the curve (AUC): GLM, 0.74 [95% confidence interval (CI): 0.65-0.83]; Naïve Bayes, 0.75 (95% CI: 0.65-0.85); SVM, 0.73 (95% CI: 0.61-0.83); and random forest, 0.72 (95% CI: 0.63-0.81). External validation was performed using a test cohort with 193 patients, and the AUC values were 0.70, 0.72, 0.73, and 0.69 for Naïve Bayes, SVM, random forest, and GLM, respectively. CONCLUSIONS: Radiomics-based machine learning approach is a very strong tool for preoperatively predicting the presence of MP/S growth patterns in lung adenocarcinoma, and can help customize treatment and surveillance strategies.

12.
Med Phys ; 48(5): 2374-2385, 2021 May.
Article in English | MEDLINE | ID: mdl-33580497

ABSTRACT

PURPOSE: The present study assessed the predictive value of peritumoral regions on three tumor tasks, and further explored the influence of peritumors with different sizes. METHODS: We retrospectively collected 333 samples of gastrointestinal stromal tumors from the Second Affiliated Hospital of Zhejiang University School of Medicine, and 183 samples of gastrointestinal stromal tumors from Tianjin Medical University Cancer Hospital. We also collected 211 samples of laryngeal carcinoma and 233 samples of nasopharyngeal carcinoma from the First Affiliated Hospital of Jinan University. The tasks of three tumor datasets were risk assessment (gastrointestinal stromal tumor), T3/T4 staging prediction (laryngeal carcinoma), and distant metastasis prediction (nasopharyngeal carcinoma), respectively. First, deep learning and radiomics were respectively used to construct peritumoral models, to study whether the peritumor had predictive value on three tumor datasets. Furthermore, we defined different sizes peritumors including fixed size (not considering tumor size) and adaptive size (according to average tumor radius) to explore the influence of peritumor of different sizes and types of tumors. Finally, we visualized the deep learning and radiomic models to observe the influence of the peritumor in three datasets. RESULTS: The performance of intra-peritumors are better than intratumors alone in three datasets. Specifically, the comparisons of area under receiver operating characteristic curve in the testing set between intra-peritumoral and intratumoral models are: 0.908 vs 0.873 (P value: 0.037) in gastrointestinal stromal tumor datasets, 0.796 vs 0.756 (P value: 0.188) in laryngeal carcinoma datasets and 0.660 vs 0.579 (P value: 0.431) in nasopharyngeal carcinoma datasets. Furthermore, for gastrointestinal stromal tumor datasets, deep learning is more stable to learn peritumors with both fixed and adaptive size than radiomics. For laryngeal carcinoma datasets, the intra-peritumoral radiomic model could make model performance more balanced. For nasopharyngeal carcinoma datasets, radiomics is also more suitable for modeling peritumors than deep learning. The size of the peritumor is critical in this task, and only the performance of 1.5 mm-4.5 mm peritumors is stable. CONCLUSIONS: Our results indicate that peritumors have additional predictive value in three tumor datasets through deep learning or radiomics. The definitions of the peritumoral region and artificial intelligence method also have great influence on the performance of the peritumor.


Subject(s)
Deep Learning , Artificial Intelligence , Humans , ROC Curve , Retrospective Studies
13.
Theranostics ; 11(5): 2098-2107, 2021.
Article in English | MEDLINE | ID: mdl-33500713

ABSTRACT

Rationale: This study aimed to use computed tomography (CT) images to assess PD-L1 expression in non-small cell lung cancer (NSCLC) and predict response to immunotherapy. Methods: We retrospectively analyzed a PD-L1 expression dataset that consisted of 939 consecutive stage IIIB-IV NSCLC patients with pretreatment CT images. A deep convolutional neural network was trained and optimized with CT images from the training cohort (n = 750) and validation cohort (n = 93) to obtain a PD-L1 expression signature (PD-L1ES), which was evaluated using the test cohort (n = 96). Finally, a separate immunotherapy cohort (n = 94) was used to assess the prognostic value of PD-L1ES with respect to clinical outcome. Results: PD-L1ES was able to predict high PD-L1 expression (PD-L1 ≥ 50%) with areas under the receiver operating characteristic curve (AUC) of 0.78 (95% confidence interval (CI): 0.75~0.80), 0.71 (95% CI: 0.59~0.81), and 0.76 (95% CI: 0.66~0.85) in the training, validation, and test cohorts, respectively. In patients treated with anti-PD-1 antibody, low PD-L1ES was associated with improved progression-free survival (PFS) (median PFS 363 days in low score group vs 183 days in high score group; hazard ratio [HR]: 2.57, 95% CI: 1.22~5.44; P = 0.010). Additionally, when PD-L1ES was combined with a clinical model that was trained using age, sex, smoking history and family history of malignancy, the response to immunotherapy could be better predicted compared to either PD-L1ES or the clinical model alone. Conclusions: The deep learning model provides a noninvasive method to predict high PD-L1 expression of NSCLC and to infer clinical outcomes in response to immunotherapy. Additionally, this deep learning model combined with clinical models demonstrated improved stratification capabilities.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/pathology , Deep Learning , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/pathology , Tomography, X-Ray Computed/methods , Aged , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Female , Humans , Immunotherapy , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Male , Middle Aged , Retrospective Studies
14.
Radiother Oncol ; 151: 1-9, 2020 10.
Article in English | MEDLINE | ID: mdl-32634460

ABSTRACT

PURPOSE: To estimate the prognostic value of deep learning (DL) magnetic resonance (MR)-based radiomics for stage T3N1M0 nasopharyngeal carcinoma (NPC) patients receiving induction chemotherapy (ICT) prior to concurrent chemoradiotherapy (CCRT). METHODS: A total of 638 stage T3N1M0 NPC patients (training cohort: n = 447; test cohort: n = 191) were enrolled and underwent MRI scans before receiving ICT + CCRT. From the pretreatment MR images, DL-based radiomic signatures were developed to predict disease-free survival (DFS) in an end-to-end way. Incorporating independent clinical prognostic parameters and radiomic signatures, a radiomic nomogram was built through multivariable Cox proportional hazards method. The discriminative performance of the radiomic nomogram was assessed using the concordance index (C-index) and the Kaplan-Meier estimator. RESULTS: Three DL-based radiomic signatures were significantly correlated with DFS in the training (C-index: 0.695-0.731, all p < 0.001) and test (C-index: 0.706-0.755, all p < 0.001) cohorts. Integrating radiomic signatures with clinical factors significantly improved the predictive value compared to the clinical model in the training (C-index: 0.771 vs. 0.640, p < 0.001) and test (C-index: 0.788 vs. 0.625, p = 0.001) cohorts. Furthermore, risk stratification using the radiomic nomogram demonstrated that the high-risk group exhibited short-lived DFS compared to the low-risk group in the training cohort (hazard ratio [HR]: 6.12, p < 0.001), which was validated in the test cohort (HR: 6.90, p < 0.001). CONCLUSIONS: Our DL-based radiomic nomogram may serve as a noninvasive and useful tool for pretreatment prognostic prediction and risk stratification in stage T3N1M0 NPC.


Subject(s)
Deep Learning , Nasopharyngeal Neoplasms , Humans , Magnetic Resonance Spectroscopy , Nasopharyngeal Carcinoma/diagnostic imaging , Nasopharyngeal Neoplasms/diagnostic imaging , Nasopharyngeal Neoplasms/radiotherapy , Nomograms , Prognosis
15.
J Immunother Cancer ; 8(2)2020 07.
Article in English | MEDLINE | ID: mdl-32636239

ABSTRACT

BACKGROUND: Tumor mutational burden (TMB) is a significant predictor of immune checkpoint inhibitors (ICIs) efficacy. This study investigated the correlation between deep learning radiomic biomarker and TMB, including its predictive value for ICIs treatment response in patients with advanced non-small-cell lung cancer (NSCLC). METHODS: CT images from 327 patients with TMB data (TMB median=6.067 mutations per megabase (range: 0 to 42.151)) were retrospectively collected and randomly divided into a training (n=236), validation (n=26), and test cohort (n=65). We used 3D-densenet to estimate the target tumor area, which used 1020 deep learning features to distinguish High-TMB from Low-TMB patients and establish the TMB radiomic biomarker (TMBRB). The TMBRB was developed in the training cohort combined with validation cohort and evaluated in the test cohort. The predictive value of TMBRB was assessed in a cohort of 123 NSCLC patients who had received ICIs (survival median=462 days (range: 16 to 1128)). RESULTS: TMBRB discriminated between High-TMB and Low-TMB patients in the training cohort (area under the curve (AUC): 0.85, 95% CI: 0.84 to 0.87))and test cohort (AUC: 0.81, 95% CI: 0.77 to 0.85). In this study, the predictive value of TMBRB was better than that of a histological subtype (AUC of training cohort: 0.75, 95% CI: 0.72 to 0.77; AUC of test cohort: 0.71, 95% CI: 0.66 to 0.76) or Radiomic model (AUC of training cohort: 0.75, 95% CI: 0.72 to 0.77; AUC of test cohort: 0.74, 95% CI: 0.69 to 0.79). When predicting immunotherapy efficacy, TMBRB divided patients into a high- and low-risk group with distinctly different overall survival (OS; HR: 0.54, 95% CI: 0.31 to 0.95; p=0.030) and progression-free survival (PFS; HR: 1.78, 95% CI: 1.07 to 2.95; p=0.023). Moreover, TMBRB had a better predictive ability when combined with the Eastern Cooperative Oncology Group performance status (OS: p=0.007; PFS: p=0.003). Visual analysis revealed that tumor microenvironment was important for predicting TMB. CONCLUSION: By combining deep learning technology and CT images, we developed an individual non-invasive biomarker that could distinguish High-TMB from Low-TMB, which might inform decisions on the use of ICIs in patients with advanced NSCLC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Immunotherapy/methods , Lung Neoplasms/drug therapy , Radiometry/methods , Female , Humans , Male , Middle Aged , Mutation , Tumor Microenvironment
16.
Eur J Radiol ; 128: 109041, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32408222

ABSTRACT

PURPOSE: To develop a deep learning-based method to assist radiologists to fast and accurately identify patients with COVID-19 by CT images. METHODS: We retrospectively collected chest CT images of 495 patients from three hospitals in China. 495 datasets were randomly divided into 395 cases (80%, 294 of COVID-19, 101 of other pneumonia) of the training set, 50 cases (10%, 37 of COVID-19, 13 of other pneumonia) of the validation set and 50 cases (10%, 37 of COVID-19, 13 of other pneumonia) of the testing set. We trained a multi-view fusion model using deep learning network to screen patients with COVID-19 using CT images with the maximum lung regions in axial, coronal and sagittal views. The performance of the proposed model was evaluated by both the validation and testing sets. RESULTS: The multi-view deep learning fusion model achieved the area under the receiver-operating characteristics curve (AUC) of 0.732, accuracy of 0.700, sensitivity of 0.730 and specificity of 0.615 in validation set. In the testing set, we can achieve AUC, accuracy, sensitivity and specificity of 0.819, 0.760, 0.811 and 0.615 respectively. CONCLUSIONS: Based on deep learning method, the proposed diagnosis model trained on multi-view images of chest CT images showed great potential to improve the efficacy of diagnosis and mitigate the heavy workload of radiologists for the initial screening of COVID-19 pneumonia.


Subject(s)
Betacoronavirus/isolation & purification , Coronavirus Infections/diagnostic imaging , Coronavirus Infections/diagnosis , Deep Learning , Mass Screening/methods , Pneumonia, Viral/diagnostic imaging , Pneumonia, Viral/diagnosis , Betacoronavirus/pathogenicity , COVID-19 , Coronavirus Infections/physiopathology , Female , Humans , Male , Middle Aged , Pandemics , Pneumonia, Viral/physiopathology , ROC Curve , Retrospective Studies , SARS-CoV-2 , Tomography, X-Ray Computed/methods
17.
J Magn Reson Imaging ; 52(4): 1102-1109, 2020 10.
Article in English | MEDLINE | ID: mdl-32212356

ABSTRACT

BACKGROUND: Gleason score (GS) is a histologic prognostic factor and the basis of treatment decision-making for prostate cancer (PCa). Treatment regimens between lower-grade (GS ≤7) and high-grade (GS >7) PCa differ largely and have great effects on cancer progression. PURPOSE: To investigate the use of different sequences in biparametric MRI (bpMRI) of the prostate gland for noninvasively distinguishing high-grade PCa. STUDY TYPE: Retrospective. POPULATION: In all, 489 patients (training cohort: N = 326; test cohort: N = 163) with PCa between June 2008 and January 2018. FIELD STRENGTH/SEQUENCE: 3.0T, pelvic phased-array coils, bpMRI including T2 -weighted imaging (T2 WI) and diffusion-weighted imaging (DWI); apparent diffusion coefficient map extracted from DWI. ASSESSMENT: The whole prostate gland was delineated. Radiomic features were extracted and selected using the Kruskal-Wallis test, the minimum redundancy-maximum relevance, and the sequential backward elimination algorithm. Two single-sequence radiomic (T2 WI, DWI) and two combined (T2 WI-DWI, T2 WI-DWI-Clinic) models were respectively constructed and validated via logistic regression. STATISTICAL TESTS: The Kruskal-Wallis test and chi-squared test were utilized to evaluate the differences among variable groups. P < 0.05 determined statistical significance. The area under the receiver operating characteristic curve (AUC), specificity, sensitivity, and accuracy were used to evaluate model performance. The Delong test was conducted to compare the differences between the AUCs of all models. RESULT: All radiomic models showed significant (P < 0.001) predictive performances. Between the single-sequence radiomic models, the DWI model achieved the most encouraging results, with AUCs of 0.801 and 0.787 in the training and test cohorts, respectively. For the combined models, the T2 WI-DWI models acquired an AUC of 0.788, which was almost the same with DWI in the test cohort, and no significant difference was found between them (training cohort: P = 0.199; test cohort: P = 0.924). DATA CONCLUSION: Radiomics based on bpMRI can noninvasively identify high-grade PCa before the operation, which is helpful for individualized diagnosis of PCa. LEVEL OF EVIDENCE: 4 TECHNICAL EFFICACY STAGE: 2 J. Magn. Reson. Imaging 2020;52:1102-1109.


Subject(s)
Magnetic Resonance Imaging , Prostatic Neoplasms , Humans , Male , Neoplasm Grading , Prostatic Neoplasms/diagnostic imaging , Retrospective Studies
18.
Br J Radiol ; 93(1108): 20190558, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31957473

ABSTRACT

OBJECTIVE: To build and validate a CT radiomic model for pre-operatively predicting lymph node metastasis in early cervical carcinoma. METHODS AND MATERIALS: A data set of 150 patients with Stage IB1 to IIA2 cervical carcinoma was retrospectively collected from the Nanfang hospital and separated into a training cohort (n = 104) and test cohort (n = 46). A total of 348 radiomic features were extracted from the delay phase of CT images. Mann-Whitney U test, recursive feature elimination, and backward elimination were used to select key radiomic features. Ridge logistics regression was used to build a radiomic model for prediction of lymph node metastasis (LNM) status by combining radiomic and clinical features. The area under the receiver operating characteristic curve (AUC) and κ test were applied to verify the model. RESULTS: Two radiomic features from delay phase CT images and one clinical feature were associated with LNM status: log-sigma-2-0 mm-3D_glcm_Idn (p = 0.01937), wavelet-HL_firstorder_Median (p = 0.03592), and Stage IB (p = 0.03608). Radiomic model was built consisting of the three features, and the AUCs were 0.80 (95% confidence interval: 0.70 ~ 0.90) and 0.75 (95% confidence intervalI: 0.53 ~ 0.93) in training and test cohorts, respectively. The κ coefficient was 0.84, showing excellent consistency. CONCLUSION: A non-invasive radiomic model, combining two radiomic features and a International Federation of Gynecology and Obstetrics stage, was built for prediction of LNM status in early cervical carcinoma. This model could serve as a pre-operative tool. ADVANCES IN KNOWLEDGE: A noninvasive CT radiomic model, combining two radiomic features and the International Federation of Gynecology and Obstetrics stage, was built for prediction of LNM status in early cervical carcinoma.


Subject(s)
Lymph Nodes/diagnostic imaging , Lymphatic Metastasis/diagnostic imaging , Tomography, X-Ray Computed , Uterine Cervical Neoplasms/diagnostic imaging , Area Under Curve , Confidence Intervals , Female , Humans , Logistic Models , Lymph Nodes/pathology , Middle Aged , Preoperative Period , ROC Curve , Retrospective Studies , Statistics, Nonparametric , Uterine Cervical Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...