Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 332
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Radiology ; 310(2): e231319, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38319168

RESUMO

Filters are commonly used to enhance specific structures and patterns in images, such as vessels or peritumoral regions, to enable clinical insights beyond the visible image using radiomics. However, their lack of standardization restricts reproducibility and clinical translation of radiomics decision support tools. In this special report, teams of researchers who developed radiomics software participated in a three-phase study (September 2020 to December 2022) to establish a standardized set of filters. The first two phases focused on finding reference filtered images and reference feature values for commonly used convolutional filters: mean, Laplacian of Gaussian, Laws and Gabor kernels, separable and nonseparable wavelets (including decomposed forms), and Riesz transformations. In the first phase, 15 teams used digital phantoms to establish 33 reference filtered images of 36 filter configurations. In phase 2, 11 teams used a chest CT image to derive reference values for 323 of 396 features computed from filtered images using 22 filter and image processing configurations. Reference filtered images and feature values for Riesz transformations were not established. Reproducibility of standardized convolutional filters was validated on a public data set of multimodal imaging (CT, fluorodeoxyglucose PET, and T1-weighted MRI) in 51 patients with soft-tissue sarcoma. At validation, reproducibility of 486 features computed from filtered images using nine configurations × three imaging modalities was assessed using the lower bounds of 95% CIs of intraclass correlation coefficients. Out of 486 features, 458 were found to be reproducible across nine teams with lower bounds of 95% CIs of intraclass correlation coefficients greater than 0.75. In conclusion, eight filter types were standardized with reference filtered images and reference feature values for verifying and calibrating radiomics software packages. A web-based tool is available for compliance checking.


Assuntos
Processamento de Imagem Assistida por Computador , Radiômica , Humanos , Reprodutibilidade dos Testes , Biomarcadores , Imagem Multimodal
2.
Artigo em Inglês | MEDLINE | ID: mdl-38861183

RESUMO

INTRODUCTION: Amyloid-ß (Aß) plaques is a significant hallmark of Alzheimer's disease (AD), detectable via amyloid-PET imaging. The Fluorine-18-Fluorodeoxyglucose ([18F]FDG) PET scan tracks cerebral glucose metabolism, correlated with synaptic dysfunction and disease progression and is complementary for AD diagnosis. Dual-scan acquisitions of amyloid PET allows the possibility to use early-phase amyloid-PET as a biomarker for neurodegeneration, proven to have a good correlation to [18F]FDG PET. The aim of this study was to evaluate the added value of synthesizing the later from the former through deep learning (DL), aiming at reducing the number of PET scans, radiation dose, and discomfort to patients. METHODS: A total of 166 subjects including cognitively unimpaired individuals (N = 72), subjects with mild cognitive impairment (N = 73) and dementia (N = 21) were included in this study. All underwent T1-weighted MRI, dual-phase amyloid PET scans using either Fluorine-18 Florbetapir ([18F]FBP) or Fluorine-18 Flutemetamol ([18F]FMM), and an [18F]FDG PET scan. Two transformer-based DL models called SwinUNETR were trained separately to synthesize the [18F]FDG from early phase [18F]FBP and [18F]FMM (eFBP/eFMM). A clinical similarity score (1: no similarity to 3: similar) was assessed to compare the imaging information obtained by synthesized [18F]FDG as well as eFBP/eFMM to actual [18F]FDG. Quantitative evaluations include region wise correlation and single-subject voxel-wise analyses in comparison with a reference [18F]FDG PET healthy control database. Dice coefficients were calculated to quantify the whole-brain spatial overlap between hypometabolic ([18F]FDG PET) and hypoperfused (eFBP/eFMM) binary maps at the single-subject level as well as between [18F]FDG PET and synthetic [18F]FDG PET hypometabolic binary maps. RESULTS: The clinical evaluation showed that, in comparison to eFBP/eFMM (average of clinical similarity score (CSS) = 1.53), the synthetic [18F]FDG images are quite similar to the actual [18F]FDG images (average of CSS = 2.7) in terms of preserving clinically relevant uptake patterns. The single-subject voxel-wise analyses showed that at the group level, the Dice scores improved by around 13% and 5% when using the DL approach for eFBP and eFMM, respectively. The correlation analysis results indicated a relatively strong correlation between eFBP/eFMM and [18F]FDG (eFBP: slope = 0.77, R2 = 0.61, P-value < 0.0001); eFMM: slope = 0.77, R2 = 0.61, P-value < 0.0001). This correlation improved for synthetic [18F]FDG (synthetic [18F]FDG generated from eFBP (slope = 1.00, R2 = 0.68, P-value < 0.0001), eFMM (slope = 0.93, R2 = 0.72, P-value < 0.0001)). CONCLUSION: We proposed a DL model for generating the [18F]FDG from eFBP/eFMM PET images. This method may be used as an alternative for multiple radiotracer scanning in research and clinical settings allowing to adopt the currently validated [18F]FDG PET normal reference databases for data analysis.

3.
Eur J Nucl Med Mol Imaging ; 51(6): 1516-1529, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38267686

RESUMO

PURPOSE: Accurate dosimetry is critical for ensuring the safety and efficacy of radiopharmaceutical therapies. In current clinical dosimetry practice, MIRD formalisms are widely employed. However, with the rapid advancement of deep learning (DL) algorithms, there has been an increasing interest in leveraging the calculation speed and automation capabilities for different tasks. We aimed to develop a hybrid transformer-based deep learning (DL) model that incorporates a multiple voxel S-value (MSV) approach for voxel-level dosimetry in [177Lu]Lu-DOTATATE therapy. The goal was to enhance the performance of the model to achieve accuracy levels closely aligned with Monte Carlo (MC) simulations, considered as the standard of reference. We extended our analysis to include MIRD formalisms (SSV and MSV), thereby conducting a comprehensive dosimetry study. METHODS: We used a dataset consisting of 22 patients undergoing up to 4 cycles of [177Lu]Lu-DOTATATE therapy. MC simulations were used to generate reference absorbed dose maps. In addition, MIRD formalism approaches, namely, single S-value (SSV) and MSV techniques, were performed. A UNEt TRansformer (UNETR) DL architecture was trained using five-fold cross-validation to generate MC-based dose maps. Co-registered CT images were fed into the network as input, whereas the difference between MC and MSV (MC-MSV) was set as output. DL results are then integrated to MSV to revive the MC dose maps. Finally, the dose maps generated by MSV, SSV, and DL were quantitatively compared to the MC reference at both voxel level and organ level (organs at risk and lesions). RESULTS: The DL approach showed slightly better performance (voxel relative absolute error (RAE) = 5.28 ± 1.32) compared to MSV (voxel RAE = 5.54 ± 1.4) and outperformed SSV (voxel RAE = 7.8 ± 3.02). Gamma analysis pass rates were 99.0 ± 1.2%, 98.8 ± 1.3%, and 98.7 ± 1.52% for DL, MSV, and SSV approaches, respectively. The computational time for MC was the highest (~2 days for a single-bed SPECT study) compared to MSV, SSV, and DL, whereas the DL-based approach outperformed the other approaches in terms of time efficiency (3 s for a single-bed SPECT). Organ-wise analysis showed absolute percent errors of 1.44 ± 3.05%, 1.18 ± 2.65%, and 1.15 ± 2.5% for SSV, MSV, and DL approaches, respectively, in lesion-absorbed doses. CONCLUSION: A hybrid transformer-based deep learning model was developed for fast and accurate dose map generation, outperforming the MIRD approaches, specifically in heterogenous regions. The model achieved accuracy close to MC gold standard and has potential for clinical implementation for use on large-scale datasets.


Assuntos
Octreotida , Octreotida/análogos & derivados , Compostos Organometálicos , Radiometria , Compostos Radiofarmacêuticos , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único , Humanos , Octreotida/uso terapêutico , Compostos Organometálicos/uso terapêutico , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único/métodos , Radiometria/métodos , Compostos Radiofarmacêuticos/uso terapêutico , Medicina de Precisão/métodos , Aprendizado Profundo , Masculino , Feminino , Método de Monte Carlo , Processamento de Imagem Assistida por Computador/métodos , Tumores Neuroendócrinos/radioterapia , Tumores Neuroendócrinos/diagnóstico por imagem
4.
Eur J Nucl Med Mol Imaging ; 51(3): 734-748, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37897616

RESUMO

PURPOSE: To investigate the impact of reduced injected doses on the quantitative and qualitative assessment of the amyloid PET tracers [18F]flutemetamol and [18F]florbetaben. METHODS: Cognitively impaired and unimpaired individuals (N = 250, 36% Aß-positive) were included and injected with [18F]flutemetamol (N = 175) or [18F]florbetaben (N = 75). PET scans were acquired in list-mode (90-110 min post-injection) and reduced-dose images were simulated to generate images of 75, 50, 25, 12.5 and 5% of the original injected dose. Images were reconstructed using vendor-provided reconstruction tools and visually assessed for Aß-pathology. SUVRs were calculated for a global cortical and three smaller regions using a cerebellar cortex reference tissue, and Centiloid was computed. Absolute and percentage differences in SUVR and CL were calculated between dose levels, and the ability to discriminate between Aß- and Aß + scans was evaluated using ROC analyses. Finally, intra-reader agreement between the reduced dose and 100% images was evaluated. RESULTS: At 5% injected dose, change in SUVR was 3.72% and 3.12%, with absolute change in Centiloid 3.35CL and 4.62CL, for [18F]flutemetamol and [18F]florbetaben, respectively. At 12.5% injected dose, percentage change in SUVR and absolute change in Centiloid were < 1.5%. AUCs for discriminating Aß- from Aß + scans were high (AUC ≥ 0.94) across dose levels, and visual assessment showed intra-reader agreement of > 80% for both tracers. CONCLUSION: This proof-of-concept study showed that for both [18F]flutemetamol and [18F]florbetaben, adequate quantitative and qualitative assessments can be obtained at 12.5% of the original injected dose. However, decisions to reduce the injected dose should be made considering the specific clinical or research circumstances.


Assuntos
Doença de Alzheimer , Compostos de Anilina , Estilbenos , Humanos , Benzotiazóis , Amiloide/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Doença de Alzheimer/diagnóstico por imagem , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo
5.
Eur J Nucl Med Mol Imaging ; 51(7): 1937-1954, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38326655

RESUMO

PURPOSE: Total metabolic tumor volume (TMTV) segmentation has significant value enabling quantitative imaging biomarkers for lymphoma management. In this work, we tackle the challenging task of automated tumor delineation in lymphoma from PET/CT scans using a cascaded approach. METHODS: Our study included 1418 2-[18F]FDG PET/CT scans from four different centers. The dataset was divided into 900 scans for development/validation/testing phases and 518 for multi-center external testing. The former consisted of 450 lymphoma, lung cancer, and melanoma scans, along with 450 negative scans, while the latter consisted of lymphoma patients from different centers with diffuse large B cell, primary mediastinal large B cell, and classic Hodgkin lymphoma cases. Our approach involves resampling PET/CT images into different voxel sizes in the first step, followed by training multi-resolution 3D U-Nets on each resampled dataset using a fivefold cross-validation scheme. The models trained on different data splits were ensemble. After applying soft voting to the predicted masks, in the second step, we input the probability-averaged predictions, along with the input imaging data, into another 3D U-Net. Models were trained with semi-supervised loss. We additionally considered the effectiveness of using test time augmentation (TTA) to improve the segmentation performance after training. In addition to quantitative analysis including Dice score (DSC) and TMTV comparisons, the qualitative evaluation was also conducted by nuclear medicine physicians. RESULTS: Our cascaded soft-voting guided approach resulted in performance with an average DSC of 0.68 ± 0.12 for the internal test data from developmental dataset, and an average DSC of 0.66 ± 0.18 on the multi-site external data (n = 518), significantly outperforming (p < 0.001) state-of-the-art (SOTA) approaches including nnU-Net and SWIN UNETR. While TTA yielded enhanced performance gains for some of the comparator methods, its impact on our cascaded approach was found to be negligible (DSC: 0.66 ± 0.16). Our approach reliably quantified TMTV, with a correlation of 0.89 with the ground truth (p < 0.001). Furthermore, in terms of visual assessment, concordance between quantitative evaluations and clinician feedback was observed in the majority of cases. The average relative error (ARE) and the absolute error (AE) in TMTV prediction on external multi-centric dataset were ARE = 0.43 ± 0.54 and AE = 157.32 ± 378.12 (mL) for all the external test data (n = 518), and ARE = 0.30 ± 0.22 and AE = 82.05 ± 99.78 (mL) when the 10% outliers (n = 53) were excluded. CONCLUSION: TMTV-Net demonstrates strong performance and generalizability in TMTV segmentation across multi-site external datasets, encompassing various lymphoma subtypes. A negligible reduction of 2% in overall performance during testing on external data highlights robust model generalizability across different centers and cancer types, likely attributable to its training with resampled inputs. Our model is publicly available, allowing easy multi-site evaluation and generalizability analysis on datasets from different institutions.


Assuntos
Processamento de Imagem Assistida por Computador , Linfoma , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Carga Tumoral , Humanos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Linfoma/diagnóstico por imagem , Processamento de Imagem Assistida por Computador/métodos , Fluordesoxiglucose F18 , Automação , Masculino , Feminino
6.
J Biomed Inform ; 150: 104583, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38191010

RESUMO

OBJECTIVE: The primary objective of our study is to address the challenge of confidentially sharing medical images across different centers. This is often a critical necessity in both clinical and research environments, yet restrictions typically exist due to privacy concerns. Our aim is to design a privacy-preserving data-sharing mechanism that allows medical images to be stored as encoded and obfuscated representations in the public domain without revealing any useful or recoverable content from the images. In tandem, we aim to provide authorized users with compact private keys that could be used to reconstruct the corresponding images. METHOD: Our approach involves utilizing a neural auto-encoder. The convolutional filter outputs are passed through sparsifying transformations to produce multiple compact codes. Each code is responsible for reconstructing different attributes of the image. The key privacy-preserving element in this process is obfuscation through the use of specific pseudo-random noise. When applied to the codes, it becomes computationally infeasible for an attacker to guess the correct representation for all the codes, thereby preserving the privacy of the images. RESULTS: The proposed framework was implemented and evaluated using chest X-ray images for different medical image analysis tasks, including classification, segmentation, and texture analysis. Additionally, we thoroughly assessed the robustness of our method against various attacks using both supervised and unsupervised algorithms. CONCLUSION: This study provides a novel, optimized, and privacy-assured data-sharing mechanism for medical images, enabling multi-party sharing in a secure manner. While we have demonstrated its effectiveness with chest X-ray images, the mechanism can be utilized in other medical images modalities as well.


Assuntos
Algoritmos , Privacidade , Disseminação de Informação
7.
J Appl Clin Med Phys ; : e14389, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38778565

RESUMO

PURPOSE: The aim of this study was to compare the organ doses assessed through a digital phantom-based and a patient specific-based dosimetric tool in adult routine thorax computed tomography (CT) examinations with reference to physical dose measurements performed in anthropomorphic phantoms. METHODS: Two Monte Carlo based dose calculation tools were used to assess organ doses in routine adult thorax CT examinations. These were a digital phantom-based dosimetry tool (NCICT, National Cancer Institute, USA) and a patient-specific individualized dosimetry tool (ImpactMC, CT Imaging GmbH, Germany). Digital phantoms and patients were classified in four groups according to their water equivalent diameter (Dw). Normalized to volume computed tomography dose index (CTDIvol), organ dose was assessed for lungs, esophagus, heart, breast, active bone marrow, and skin. Organ doses were compared to measurements performed using thermoluminescent detectors (TLDs) in two physical anthropomorphic phantoms that simulate the average adult individual as a male (Alderson Research Labs, USA) and as a female (ATOM Phantoms, USA). RESULTS: The average percent difference of NCICT to TLD and ImpactMC to TLD dose measurements across all organs in both sexes was 13% and 6%, respectively. The average ± 1 standard deviation in dose values across all organs with NCICT, ImpactMC, and TLDs was ± 0.06 (mGy/mGy), ± 0.19 (mGy/mGy), and ± 0.13 (mGy/mGy), respectively. Organ doses decreased with increasing Dw in both NCICT and ImpactMC. CONCLUSION: Organ doses estimated with ImpactMC were in closer agreement to TLDs compared to NCICT. This may be attributed to the inherent property of ImpactMC methodology to generate phantoms that resemble the realistic anatomy of the examined patient as opposed to NCICT methodology that incorporates an anatomical discrepancy between phantoms and patients.

8.
Eur J Nucl Med Mol Imaging ; 50(10): 2984-2996, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37171633

RESUMO

PURPOSE: Metastatic neuroendocrine tumors (NETs) overexpressing type 2 somatostatin receptors are the target for peptide receptor radionuclide therapy (PRRT) through the theragnostic pair of 68Ga/177Lu-DOTATATE. The main purpose of this study was to develop machine learning models to predict therapeutic tumor dose using pre therapy 68Ga -PET and clinicopathological biomarkers. METHODS: We retrospectively analyzed 90 segmented metastatic NETs from 25 patients (M14/F11, age 63.7 ± 9.5, range 38-76) treated by 177Lu-DOTATATE at our institute. Patients underwent both pretherapy [68Ga]Ga-DOTA-TATE PET/CT and four timepoints SPECT/CT at ~ 4, 24, 96, and 168 h post-177Lu-DOTATATE infusion. Tumors were segmented by a radiologist on baseline CT or MRI and transferred to co-registered PET/CT and SPECT/CT, and normal organs were segmented by deep learning-based method on CT of the PET and SPECT. The SUV metrics and tumor-to-normal tissue SUV ratios (SUV_TNRs) were calculated from 68Ga -PET at the contour-level. Posttherapy dosimetry was performed based on the co-registration of SPECT/CTs to generate time-integrated-activity, followed by an in-house Monte Carlo-based absorbed dose estimation. The correlation between delivered 177Lu Tumor absorbed dose and PET-derived metrics along with baseline clinicopathological biomarkers (such as Creatinine, Chromogranin A and prior therapies) were evaluated. Multiple interpretable machine-learning algorithms were developed to predict tumor dose using these pretherapy information. Model performance on a nested tenfold cross-validation was evaluated in terms of coefficient of determination (R2), mean-absolute-error (MAE), and mean-relative-absolute-error (MRAE). RESULTS: SUVmean showed a significant correlation (q-value < 0.05) with absorbed dose (Spearman ρ = 0.64), followed by TLSUVmean (SUVmean of total-lesion-burden) and SUVpeak (ρ = 0.45 and 0.41, respectively). The predictive value of PET-SUVmean in estimation of posttherapy absorbed dose was stronger compared to PET-SUVpeak, and SUV_TNRs in terms of univariate analysis (R2 = 0.28 vs. R2 ≤ 0.12). An optimal trivariate random forest model composed of SUVmean, TLSUVmean, and total liver SUVmean (normal and tumoral liver) provided the best performance in tumor dose prediction with R2 = 0.64, MAE = 0.73 Gy/GBq, and MRAE = 0.2. CONCLUSION: Our preliminary results demonstrate the feasibility of using baseline PET images for prediction of absorbed dose prior to 177Lu-PRRT. Machine learning models combining multiple PET-based metrics performed better than using a single SUV value and using other investigated clinicopathological biomarkers. Developing such quantitative models forms the groundwork for the role of 68Ga -PET not only for the implementation of personalized treatment planning but also for patient stratification in the era of precision medicine.


Assuntos
Tumores Neuroendócrinos , Compostos Organometálicos , Humanos , Pessoa de Meia-Idade , Idoso , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Radioisótopos de Gálio , Octreotida/uso terapêutico , Estudos Retrospectivos , Compostos Organometálicos/uso terapêutico , Tumores Neuroendócrinos/diagnóstico por imagem , Tumores Neuroendócrinos/radioterapia , Tumores Neuroendócrinos/tratamento farmacológico , Biomarcadores
9.
Eur J Nucl Med Mol Imaging ; 50(7): 1881-1896, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36808000

RESUMO

PURPOSE: Partial volume effect (PVE) is a consequence of the limited spatial resolution of PET scanners. PVE can cause the intensity values of a particular voxel to be underestimated or overestimated due to the effect of surrounding tracer uptake. We propose a novel partial volume correction (PVC) technique to overcome the adverse effects of PVE on PET images. METHODS: Two hundred and twelve clinical brain PET scans, including 50 18F-Fluorodeoxyglucose (18F-FDG), 50 18F-Flortaucipir, 36 18F-Flutemetamol, and 76 18F-FluoroDOPA, and their corresponding T1-weighted MR images were enrolled in this study. The Iterative Yang technique was used for PVC as a reference or surrogate of the ground truth for evaluation. A cycle-consistent adversarial network (CycleGAN) was trained to directly map non-PVC PET images to PVC PET images. Quantitative analysis using various metrics, including structural similarity index (SSIM), root mean squared error (RMSE), and peak signal-to-noise ratio (PSNR), was performed. Furthermore, voxel-wise and region-wise-based correlations of activity concentration between the predicted and reference images were evaluated through joint histogram and Bland and Altman analysis. In addition, radiomic analysis was performed by calculating 20 radiomic features within 83 brain regions. Finally, a voxel-wise two-sample t-test was used to compare the predicted PVC PET images with reference PVC images for each radiotracer. RESULTS: The Bland and Altman analysis showed the largest and smallest variance for 18F-FDG (95% CI: - 0.29, + 0.33 SUV, mean = 0.02 SUV) and 18F-Flutemetamol (95% CI: - 0.26, + 0.24 SUV, mean = - 0.01 SUV), respectively. The PSNR was lowest (29.64 ± 1.13 dB) for 18F-FDG and highest (36.01 ± 3.26 dB) for 18F-Flutemetamol. The smallest and largest SSIM were achieved for 18F-FDG (0.93 ± 0.01) and 18F-Flutemetamol (0.97 ± 0.01), respectively. The average relative error for the kurtosis radiomic feature was 3.32%, 9.39%, 4.17%, and 4.55%, while it was 4.74%, 8.80%, 7.27%, and 6.81% for NGLDM_contrast feature for 18F-Flutemetamol, 18F-FluoroDOPA, 18F-FDG, and 18F-Flortaucipir, respectively. CONCLUSION: An end-to-end CycleGAN PVC method was developed and evaluated. Our model generates PVC images from the original non-PVC PET images without requiring additional anatomical information, such as MRI or CT. Our model eliminates the need for accurate registration or segmentation or PET scanner system response characterization. In addition, no assumptions regarding anatomical structure size, homogeneity, boundary, or background level are required.


Assuntos
Compostos de Anilina , Fluordesoxiglucose F18 , Humanos , Tomografia por Emissão de Pósitrons/métodos , Encéfalo/diagnóstico por imagem , Processamento de Imagem Assistida por Computador/métodos
10.
Eur J Nucl Med Mol Imaging ; 50(4): 1034-1050, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36508026

RESUMO

PURPOSE: Attenuation correction and scatter compensation (AC/SC) are two main steps toward quantitative PET imaging, which remain challenging in PET-only and PET/MRI systems. These can be effectively tackled via deep learning (DL) methods. However, trustworthy, and generalizable DL models commonly require well-curated, heterogeneous, and large datasets from multiple clinical centers. At the same time, owing to legal/ethical issues and privacy concerns, forming a large collective, centralized dataset poses significant challenges. In this work, we aimed to develop a DL-based model in a multicenter setting without direct sharing of data using federated learning (FL) for AC/SC of PET images. METHODS: Non-attenuation/scatter corrected and CT-based attenuation/scatter corrected (CT-ASC) 18F-FDG PET images of 300 patients were enrolled in this study. The dataset consisted of 6 different centers, each with 50 patients, with scanner, image acquisition, and reconstruction protocols varying across the centers. CT-based ASC PET images served as the standard reference. All images were reviewed to include high-quality and artifact-free PET images. Both corrected and uncorrected PET images were converted to standardized uptake values (SUVs). We used a modified nested U-Net utilizing residual U-block in a U-shape architecture. We evaluated two FL models, namely sequential (FL-SQ) and parallel (FL-PL) and compared their performance with the baseline centralized (CZ) learning model wherein the data were pooled to one server, as well as center-based (CB) models where for each center the model was built and evaluated separately. Data from each center were divided to contribute to training (30 patients), validation (10 patients), and test sets (10 patients). Final evaluations and reports were performed on 60 patients (10 patients from each center). RESULTS: In terms of percent SUV absolute relative error (ARE%), both FL-SQ (CI:12.21-14.81%) and FL-PL (CI:11.82-13.84%) models demonstrated excellent agreement with the centralized framework (CI:10.32-12.00%), while FL-based algorithms improved model performance by over 11% compared to CB training strategy (CI: 22.34-26.10%). Furthermore, the Mann-Whitney test between different strategies revealed no significant differences between CZ and FL-based algorithms (p-value > 0.05) in center-categorized mode. At the same time, a significant difference was observed between the different training approaches on the overall dataset (p-value < 0.05). In addition, voxel-wise comparison, with respect to reference CT-ASC, exhibited similar performance for images predicted by CZ (R2 = 0.94), FL-SQ (R2 = 0.93), and FL-PL (R2 = 0.92), while CB model achieved a far lower coefficient of determination (R2 = 0.74). Despite the strong correlations between CZ and FL-based methods compared to reference CT-ASC, a slight underestimation of predicted voxel values was observed. CONCLUSION: Deep learning-based models provide promising results toward quantitative PET image reconstruction. Specifically, we developed two FL models and compared their performance with center-based and centralized models. The proposed FL-based models achieved higher performance compared to center-based models, comparable with centralized models. Our work provided strong empirical evidence that the FL framework can fully benefit from the generalizability and robustness of DL models used for AC/SC in PET, while obviating the need for the direct sharing of datasets between clinical imaging centers.


Assuntos
Aprendizado Profundo , Processamento de Imagem Assistida por Computador , Humanos , Processamento de Imagem Assistida por Computador/métodos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Tomografia por Emissão de Pósitrons/métodos , Imageamento por Ressonância Magnética/métodos
11.
Eur J Nucl Med Mol Imaging ; 51(1): 40-53, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37682303

RESUMO

PURPOSE: Image artefacts continue to pose challenges in clinical molecular imaging, resulting in misdiagnoses, additional radiation doses to patients and financial costs. Mismatch and halo artefacts occur frequently in gallium-68 (68Ga)-labelled compounds whole-body PET/CT imaging. Correcting for these artefacts is not straightforward and requires algorithmic developments, given that conventional techniques have failed to address them adequately. In the current study, we employed differential privacy-preserving federated transfer learning (FTL) to manage clinical data sharing and tackle privacy issues for building centre-specific models that detect and correct artefacts present in PET images. METHODS: Altogether, 1413 patients with 68Ga prostate-specific membrane antigen (PSMA)/DOTA-TATE (TOC) PET/CT scans from 3 countries, including 8 different centres, were enrolled in this study. CT-based attenuation and scatter correction (CT-ASC) was used in all centres for quantitative PET reconstruction. Prior to model training, an experienced nuclear medicine physician reviewed all images to ensure the use of high-quality, artefact-free PET images (421 patients' images). A deep neural network (modified U2Net) was trained on 80% of the artefact-free PET images to utilize centre-based (CeBa), centralized (CeZe) and the proposed differential privacy FTL frameworks. Quantitative analysis was performed in 20% of the clean data (with no artefacts) in each centre. A panel of two nuclear medicine physicians conducted qualitative assessment of image quality, diagnostic confidence and image artefacts in 128 patients with artefacts (256 images for CT-ASC and FTL-ASC). RESULTS: The three approaches investigated in this study for 68Ga-PET imaging (CeBa, CeZe and FTL) resulted in a mean absolute error (MAE) of 0.42 ± 0.21 (CI 95%: 0.38 to 0.47), 0.32 ± 0.23 (CI 95%: 0.27 to 0.37) and 0.28 ± 0.15 (CI 95%: 0.25 to 0.31), respectively. Statistical analysis using the Wilcoxon test revealed significant differences between the three approaches, with FTL outperforming CeBa and CeZe (p-value < 0.05) in the clean test set. The qualitative assessment demonstrated that FTL-ASC significantly improved image quality and diagnostic confidence and decreased image artefacts, compared to CT-ASC in 68Ga-PET imaging. In addition, mismatch and halo artefacts were successfully detected and disentangled in the chest, abdomen and pelvic regions in 68Ga-PET imaging. CONCLUSION: The proposed approach benefits from using large datasets from multiple centres while preserving patient privacy. Qualitative assessment by nuclear medicine physicians showed that the proposed model correctly addressed two main challenging artefacts in 68Ga-PET imaging. This technique could be integrated in the clinic for 68Ga-PET imaging artefact detection and disentanglement using multicentric heterogeneous datasets.


Assuntos
Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Neoplasias da Próstata , Masculino , Humanos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Artefatos , Radioisótopos de Gálio , Privacidade , Tomografia por Emissão de Pósitrons/métodos , Aprendizado de Máquina , Processamento de Imagem Assistida por Computador/métodos
12.
Eur Radiol ; 33(12): 9411-9424, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37368113

RESUMO

OBJECTIVE: We propose a deep learning-guided approach to generate voxel-based absorbed dose maps from whole-body CT acquisitions. METHODS: The voxel-wise dose maps corresponding to each source position/angle were calculated using Monte Carlo (MC) simulations considering patient- and scanner-specific characteristics (SP_MC). The dose distribution in a uniform cylinder was computed through MC calculations (SP_uniform). The density map and SP_uniform dose maps were fed into a residual deep neural network (DNN) to predict SP_MC through an image regression task. The whole-body dose maps reconstructed by the DNN and MC were compared in the 11 test cases scanned with two tube voltages through transfer learning with/without tube current modulation (TCM). The voxel-wise and organ-wise dose evaluations, such as mean error (ME, mGy), mean absolute error (MAE, mGy), relative error (RE, %), and relative absolute error (RAE, %), were performed. RESULTS: The model performance for the 120 kVp and TCM test set in terms of ME, MAE, RE, and RAE voxel-wise parameters was - 0.0302 ± 0.0244 mGy, 0.0854 ± 0.0279 mGy, - 1.13 ± 1.41%, and 7.17 ± 0.44%, respectively. The organ-wise errors for 120 kVp and TCM scenario averaged over all segmented organs in terms of ME, MAE, RE, and RAE were - 0.144 ± 0.342 mGy, and 0.23 ± 0.28 mGy, - 1.11 ± 2.90%, 2.34 ± 2.03%, respectively. CONCLUSION: Our proposed deep learning model is able to generate voxel-level dose maps from a whole-body CT scan with reasonable accuracy suitable for organ-level absorbed dose estimation. CLINICAL RELEVANCE STATEMENT: We proposed a novel method for voxel dose map calculation using deep neural networks. This work is clinically relevant since accurate dose calculation for patients can be carried out within acceptable computational time compared to lengthy Monte Carlo calculations. KEY POINTS: • We proposed a deep neural network approach as an alternative to Monte Carlo dose calculation. • Our proposed deep learning model is able to generate voxel-level dose maps from a whole-body CT scan with reasonable accuracy, suitable for organ-level dose estimation. • By generating a dose distribution from a single source position, our model can generate accurate and personalized dose maps for a wide range of acquisition parameters.


Assuntos
Redes Neurais de Computação , Imagem Corporal Total , Humanos , Imagens de Fantasmas , Método de Monte Carlo , Tomografia Computadorizada por Raios X , Doses de Radiação
13.
Eur Radiol ; 33(5): 3243-3252, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36703015

RESUMO

OBJECTIVES: This study aimed to improve patient positioning accuracy by relying on a CT localizer and a deep neural network to optimize image quality and radiation dose. METHODS: We included 5754 chest CT axial and anterior-posterior (AP) images from two different centers, C1 and C2. After pre-processing, images were split into training (80%) and test (20%) datasets. A deep neural network was trained to generate 3D axial images from the AP localizer. The geometric centerlines of patient bodies were indicated by creating a bounding box on the predicted images. The distance between the body centerline, estimated by the deep learning model and ground truth (BCAP), was compared with patient mis-centering during manual positioning (BCMP). We evaluated the performance of our model in terms of distance between the lung centerline estimated by the deep learning model and the ground truth (LCAP). RESULTS: The error in terms of BCAP was - 0.75 ± 7.73 mm and 2.06 ± 10.61 mm for C1 and C2, respectively. This error was significantly lower than BCMP, which achieved an error of 9.35 ± 14.94 and 13.98 ± 14.5 mm for C1 and C2, respectively. The absolute BCAP was 5.7 ± 5.26 and 8.26 ± 6.96 mm for C1 and C2, respectively. The LCAP metric was 1.56 ± 10.8 and -0.27 ± 16.29 mm for C1 and C2, respectively. The error in terms of BCAP and LCAP was higher for larger patients (p value < 0.01). CONCLUSION: The accuracy of the proposed method was comparable to available alternative methods, carrying the advantage of being free from errors related to objects blocking the camera visibility. KEY POINTS: • Patient mis-centering in the anterior-posterior direction (AP) is a common problem in clinical practice which can degrade image quality and increase patient radiation dose. • We proposed a deep neural network for automatic patient positioning using only the CT image localizer, achieving a performance comparable to alternative techniques, such as the external 3D visual camera. • The advantage of the proposed method is that it is free from errors related to objects blocking the camera visibility and that it could be implemented on imaging consoles as a patient positioning support tool.


Assuntos
Redes Neurais de Computação , Tomografia Computadorizada por Raios X , Humanos , Tomografia Computadorizada por Raios X/métodos , Imageamento Tridimensional , Posicionamento do Paciente/métodos , Processamento de Imagem Assistida por Computador/métodos
14.
Radiol Med ; 128(12): 1521-1534, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37751102

RESUMO

PURPOSE: Glioblastoma Multiforme (GBM) represents the predominant aggressive primary tumor of the brain with short overall survival (OS) time. We aim to assess the potential of radiomic features in predicting the time-to-event OS of patients with GBM using machine learning (ML) algorithms. MATERIALS AND METHODS: One hundred nineteen patients with GBM, who had T1-weighted contrast-enhanced and T2-FLAIR MRI sequences, along with clinical data and survival time, were enrolled. Image preprocessing methods included 64 bin discretization, Laplacian of Gaussian (LOG) filters with three Sigma values and eight variations of Wavelet Transform. Images were then segmented, followed by the extraction of 1212 radiomic features. Seven feature selection (FS) methods and six time-to-event ML algorithms were utilized. The combination of preprocessing, FS, and ML algorithms (12 × 7 × 6 = 504 models) was evaluated by multivariate analysis. RESULTS: Our multivariate analysis showed that the best prognostic FS/ML combinations are the Mutual Information (MI)/Cox Boost, MI/Generalized Linear Model Boosting (GLMB) and MI/Generalized Linear Model Network (GLMN), all of which were done via the LOG (Sigma = 1 mm) preprocessing method (C-index = 0.77). The LOG filter with Sigma = 1 mm preprocessing method, MI, GLMB and GLMN achieved significantly higher C-indices than other preprocessing, FS, and ML methods (all p values < 0.05, mean C-indices of 0.65, 0.70, and 0.64, respectively). CONCLUSION: ML algorithms are capable of predicting the time-to-event OS of patients using MRI-based radiomic and clinical features. MRI-based radiomics analysis in combination with clinical variables might appear promising in assisting clinicians in the survival prediction of patients with GBM. Further research is needed to establish the applicability of radiomics in the management of GBM in the clinic.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/patologia , Imageamento por Ressonância Magnética/métodos , Encéfalo/patologia , Prognóstico , Proteínas Adaptadoras de Transdução de Sinal
15.
J Digit Imaging ; 36(6): 2494-2506, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37735309

RESUMO

Heart failure caused by iron deposits in the myocardium is the primary cause of mortality in beta-thalassemia major patients. Cardiac magnetic resonance imaging (CMRI) T2* is the primary screening technique used to detect myocardial iron overload, but inherently bears some limitations. In this study, we aimed to differentiate beta-thalassemia major patients with myocardial iron overload from those without myocardial iron overload (detected by T2*CMRI) based on radiomic features extracted from echocardiography images and machine learning (ML) in patients with normal left ventricular ejection fraction (LVEF > 55%) in echocardiography. Out of 91 cases, 44 patients with thalassemia major with normal LVEF (> 55%) and T2* ≤ 20 ms and 47 people with LVEF > 55% and T2* > 20 ms as the control group were included in the study. Radiomic features were extracted for each end-systolic (ES) and end-diastolic (ED) image. Then, three feature selection (FS) methods and six different classifiers were used. The models were evaluated using various metrics, including the area under the ROC curve (AUC), accuracy (ACC), sensitivity (SEN), and specificity (SPE). Maximum relevance-minimum redundancy-eXtreme gradient boosting (MRMR-XGB) (AUC = 0.73, ACC = 0.73, SPE = 0.73, SEN = 0.73), ANOVA-MLP (AUC = 0.69, ACC = 0.69, SPE = 0.56, SEN = 0.83), and recursive feature elimination-K-nearest neighbors (RFE-KNN) (AUC = 0.65, ACC = 0.65, SPE = 0.64, SEN = 0.65) were the best models in ED, ES, and ED&ES datasets. Using radiomic features extracted from echocardiographic images and ML, it is feasible to predict cardiac problems caused by iron overload.


Assuntos
Sobrecarga de Ferro , Talassemia , Disfunção Ventricular Esquerda , Talassemia beta , Humanos , Talassemia beta/complicações , Talassemia beta/diagnóstico por imagem , Volume Sistólico , Função Ventricular Esquerda , Talassemia/complicações , Talassemia/diagnóstico por imagem , Miocárdio , Ecocardiografia/métodos , Sobrecarga de Ferro/complicações , Sobrecarga de Ferro/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Disfunção Ventricular Esquerda/etiologia , Disfunção Ventricular Esquerda/complicações
16.
J Digit Imaging ; 36(4): 1348-1363, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37059890

RESUMO

In this study, the ability of radiomics features extracted from myocardial perfusion imaging with SPECT (MPI-SPECT) was investigated for the prediction of ejection fraction (EF) post-percutaneous coronary intervention (PCI) treatment. A total of 52 patients who had undergone pre-PCI MPI-SPECT were enrolled in this study. After normalization of the images, features were extracted from the left ventricle, initially automatically segmented by k-means and active contour methods, and finally edited and approved by an expert radiologist. More than 1700 2D and 3D radiomics features were extracted from each patient's scan. A cross-combination of three feature selections and seven classifier methods was implemented. Three classes of no or dis-improvement (class 1), improved EF from 0 to 5% (class 2), and improved EF over 5% (class 3) were predicted by using tenfold cross-validation. Lastly, the models were evaluated based on accuracy, AUC, sensitivity, specificity, precision, and F-score. Neighborhood component analysis (NCA) selected the most predictive feature signatures, including Gabor, first-order, and NGTDM features. Among the classifiers, the best performance was achieved by the fine KNN classifier, which yielded mean accuracy, AUC, sensitivity, specificity, precision, and F-score of 0.84, 0.83, 0.75, 0.87, 0.78, and 0.76, respectively, in 100 iterations of classification, within the 52 patients with 10-fold cross-validation. The MPI-SPECT-based radiomic features are well suited for predicting post-revascularization EF and therefore provide a helpful approach for deciding on the most appropriate treatment.


Assuntos
Imagem de Perfusão do Miocárdio , Intervenção Coronária Percutânea , Humanos , Volume Sistólico , Tomografia Computadorizada de Emissão de Fóton Único , Aprendizado de Máquina , Perfusão
17.
J Digit Imaging ; 36(2): 497-509, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36376780

RESUMO

A U-shaped contraction pattern was shown to be associated with a better Cardiac resynchronization therapy (CRT) response. The main goal of this study is to automatically recognize left ventricular contractile patterns using machine learning algorithms trained on conventional quantitative features (ConQuaFea) and radiomic features extracted from Gated single-photon emission computed tomography myocardial perfusion imaging (GSPECT MPI). Among 98 patients with standard resting GSPECT MPI included in this study, 29 received CRT therapy and 69 did not (also had CRT inclusion criteria but did not receive treatment yet at the time of data collection, or refused treatment). A total of 69 non-CRT patients were employed for training, and the 29 were employed for testing. The models were built utilizing features from three distinct feature sets (ConQuaFea, radiomics, and ConQuaFea + radiomics (combined)), which were chosen using Recursive feature elimination (RFE) feature selection (FS), and then trained using seven different machine learning (ML) classifiers. In addition, CRT outcome prediction was assessed by different treatment inclusion criteria as the study's final phase. The MLP classifier had the highest performance among ConQuaFea models (AUC, SEN, SPE = 0.80, 0.85, 0.76). RF achieved the best performance in terms of AUC, SEN, and SPE with values of 0.65, 0.62, and 0.68, respectively, among radiomic models. GB and RF approaches achieved the best AUC, SEN, and SPE values of 0.78, 0.92, and 0.63 and 0.74, 0.93, and 0.56, respectively, among the combined models. A promising outcome was obtained when using radiomic and ConQuaFea from GSPECT MPI to detect left ventricular contractile patterns by machine learning.


Assuntos
Imagem de Perfusão do Miocárdio , Humanos , Tomografia Computadorizada de Emissão de Fóton Único , Aprendizado de Máquina , Algoritmos , Perfusão
18.
Hum Brain Mapp ; 43(16): 5032-5043, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36087092

RESUMO

We aim to synthesize brain time-of-flight (TOF) PET images/sinograms from their corresponding non-TOF information in the image space (IS) and sinogram space (SS) to increase the signal-to-noise ratio (SNR) and contrast of abnormalities, and decrease the bias in tracer uptake quantification. One hundred forty clinical brain 18 F-FDG PET/CT scans were collected to generate TOF and non-TOF sinograms. The TOF sinograms were split into seven time bins (0, ±1, ±2, ±3). The predicted TOF sinogram was reconstructed and the performance of both models (IS and SS) compared with reference TOF and non-TOF. Wide-ranging quantitative and statistical analysis metrics, including structural similarity index metric (SSIM), root mean square error (RMSE), as well as 28 radiomic features for 83 brain regions were extracted to evaluate the performance of the CycleGAN model. SSIM and RMSE of 0.99 ± 0.03, 0.98 ± 0.02 and 0.12 ± 0.09, 0.16 ± 0.04 were achieved for the generated TOF-PET images in IS and SS, respectively. They were 0.97 ± 0.03 and 0.22 ± 0.12, respectively, for non-TOF-PET images. The Bland & Altman analysis revealed that the lowest tracer uptake value bias (-0.02%) and minimum variance (95% CI: -0.17%, +0.21%) were achieved for TOF-PET images generated in IS. For malignant lesions, the contrast in the test dataset was enhanced from 3.22 ± 2.51 for non-TOF to 3.34 ± 0.41 and 3.65 ± 3.10 for TOF PET in SS and IS, respectively. The implemented CycleGAN is capable of generating TOF from non-TOF PET images to achieve better image quality.


Assuntos
Aprendizado Profundo , Fluordesoxiglucose F18 , Humanos , Processamento de Imagem Assistida por Computador/métodos , Tomografia por Emissão de Pósitrons , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Encéfalo/diagnóstico por imagem
19.
Magn Reson Med ; 87(2): 686-701, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34480771

RESUMO

PURPOSE: We compare the performance of three commonly used MRI-guided attenuation correction approaches in torso PET/MRI, namely segmentation-, atlas-, and deep learning-based algorithms. METHODS: Twenty-five co-registered torso 18 F-FDG PET/CT and PET/MR images were enrolled. PET attenuation maps were generated from in-phase Dixon MRI using a three-tissue class segmentation-based approach (soft-tissue, lung, and background air), voxel-wise weighting atlas-based approach, and a residual convolutional neural network. The bias in standardized uptake value (SUV) was calculated for each approach considering CT-based attenuation corrected PET images as reference. In addition to the overall performance assessment of these approaches, the primary focus of this work was on recognizing the origins of potential outliers, notably body truncation, metal-artifacts, abnormal anatomy, and small malignant lesions in the lungs. RESULTS: The deep learning approach outperformed both atlas- and segmentation-based methods resulting in less than 4% SUV bias across 25 patients compared to the segmentation-based method with up to 20% SUV bias in bony structures and the atlas-based method with 9% bias in the lung. The deep learning-based method exhibited superior performance. Yet, in case of sever truncation and metallic-artifacts in the input MRI, this approach was outperformed by the atlas-based method, exhibiting suboptimal performance in the affected regions. Conversely, for abnormal anatomies, such as a patient presenting with one lung or small malignant lesion in the lung, the deep learning algorithm exhibited promising performance compared to other methods. CONCLUSION: The deep learning-based method provides promising outcome for synthetic CT generation from MRI. However, metal-artifact and body truncation should be specifically addressed.


Assuntos
Aprendizado Profundo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Algoritmos , Humanos , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Tomografia por Emissão de Pósitrons , Tronco
20.
Annu Rev Biomed Eng ; 23: 249-276, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-33797938

RESUMO

The widespread availability of high-performance computing and the popularity of artificial intelligence (AI) with machine learning and deep learning (ML/DL) algorithms at the helm have stimulated the development of many applications involving the use of AI-based techniques in molecular imaging research. Applications reported in the literature encompass various areas, including innovative design concepts in positron emission tomography (PET) instrumentation, quantitative image reconstruction and analysis techniques, computer-aided detection and diagnosis, as well as modeling and prediction of outcomes. This review reflects the tremendous interest in quantitative molecular imaging using ML/DL techniques during the past decade, ranging from the basic principles of ML/DL techniques to the various steps required for obtaining quantitatively accurate PET data, including algorithms used to denoise or correct for physical degrading factors as well as to quantify tracer uptake and metabolic tumor volume for treatment monitoring or radiation therapy treatment planning and response prediction.This review also addresses future opportunities and current challenges facing the adoption of ML/DL approaches and their role in multimodality imaging.


Assuntos
Inteligência Artificial , Aprendizado Profundo , Algoritmos , Humanos , Processamento de Imagem Assistida por Computador , Tomografia por Emissão de Pósitrons
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa