Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 334
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38981950

RESUMO

BACKGROUND: Overall Survival (OS) and Progression-Free Survival (PFS) analyses are crucial metrics for evaluating the efficacy and impact of treatment. This study evaluated the role of clinical biomarkers and dosimetry parameters on survival outcomes of patients undergoing 90Y selective internal radiation therapy (SIRT). MATERIALS/METHODS: This preliminary and retrospective analysis included 17 patients with hepatocellular carcinoma (HCC) treated with 90Y SIRT. The patients underwent personalized treatment planning and voxel-wise dosimetry. After the procedure, the OS and PFS were evaluated. Three structures were delineated including tumoral liver (TL), normal perfused liver (NPL), and whole normal liver (WNL). 289 dose-volume constraints (DVCs) were extracted from dose-volume histograms of physical and biological effective dose (BED) maps calculated on 99mTc-MAA and 90Y SPECT/CT images. Subsequently, the DVCs and 16 clinical biomarkers were used as features for univariate and multivariate analysis. Cox proportional hazard ratio (HR) was employed for univariate analysis. HR and the concordance index (C-Index) were calculated for each feature. Using eight different strategies, a cross-combination of various models and feature selection (FS) methods was applied for multivariate analysis. The performance of each model was assessed using an averaged C-Index on a three-fold nested cross-validation framework. The Kaplan-Meier (KM) curve was employed for univariate and machine learning (ML) model performance assessment. RESULTS: The median OS was 11 months [95% CI: 8.5, 13.09], whereas the PFS was seven months [95% CI: 5.6, 10.98]. Univariate analysis demonstrated the presence of Ascites (HR: 9.2[1.8,47]) and the aim of SIRT (segmentectomy, lobectomy, palliative) (HR: 0.066 [0.0057, 0.78]), Aspartate aminotransferase (AST) level (HR:0.1 [0.012-0.86]), and MAA-Dose-V205(%)-TL (HR:8.5[1,72]) as predictors for OS. 90Y-derived parameters were associated with PFS but not with OS. MAA-Dose-V205(%)-WNL, MAA-BED-V400(%)-WNL with (HR:13 [1.5-120]) and 90Y-Dose-mean-TL, 90Y-D50-TL-Gy, 90Y-Dose-V205(%)-TL, 90Y-Dose- D50-TL-Gy, and 90Y-BED-V400(%)-TL (HR:15 [1.8-120]) were highly associated with PFS among dosimetry parameters. The highest C-index observed in multivariate analysis using ML was 0.94 ± 0.13 obtained from Variable Hunting-variable-importance (VH.VIMP) FS and Cox Proportional Hazard model predicting OS, using clinical features. However, the combination of VH. VIMP FS method with a Generalized Linear Model Network model predicting OS using Therapy strategy features outperformed the other models in terms of both C-index and stratification of KM curves (C-Index: 0.93 ± 0.14 and log-rank p-value of 0.023 for KM curve stratification). CONCLUSION: This preliminary study confirmed the role played by baseline clinical biomarkers and dosimetry parameters in predicting the treatment outcome, paving the way for the establishment of a dose-effect relationship. In addition, the feasibility of using ML along with these features was demonstrated as a helpful tool in the clinical management of patients, both prior to and following 90Y-SIRT.

2.
Theranostics ; 14(9): 3404-3422, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38948052

RESUMO

Radiopharmaceutical therapy (RPT) is a rapidly developing field of nuclear medicine, with several RPTs already well established in the treatment of several different types of cancers. However, the current approaches to RPTs often follow a somewhat inflexible "one size fits all" paradigm, where patients are administered the same amount of radioactivity per cycle regardless of their individual characteristics and features. This approach fails to consider inter-patient variations in radiopharmacokinetics, radiation biology, and immunological factors, which can significantly impact treatment outcomes. To address this limitation, we propose the development of theranostic digital twins (TDTs) to personalize RPTs based on actual patient data. Our proposed roadmap outlines the steps needed to create and refine TDTs that can optimize radiation dose to tumors while minimizing toxicity to organs at risk. The TDT models incorporate physiologically-based radiopharmacokinetic (PBRPK) models, which are additionally linked to a radiobiological optimizer and an immunological modulator, taking into account factors that influence RPT response. By using TDT models, we envisage the ability to perform virtual clinical trials, selecting therapies towards improved treatment outcomes while minimizing risks associated with secondary effects. This framework could empower practitioners to ultimately develop tailored RPT solutions for subgroups and individual patients, thus improving the precision, accuracy, and efficacy of treatments while minimizing risks to patients. By incorporating TDT models into RPTs, we can pave the way for a new era of precision medicine in cancer treatment.


Assuntos
Neoplasias , Medicina de Precisão , Compostos Radiofarmacêuticos , Humanos , Medicina de Precisão/métodos , Neoplasias/terapia , Neoplasias/radioterapia , Compostos Radiofarmacêuticos/uso terapêutico , Compostos Radiofarmacêuticos/farmacocinética
3.
Theranostics ; 14(9): 3708-3718, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38948061

RESUMO

Purpose: This study aims to elucidate the role of quantitative SSTR-PET metrics and clinicopathological biomarkers in the progression-free survival (PFS) and overall survival (OS) of neuroendocrine tumors (NETs) treated with peptide receptor radionuclide therapy (PRRT). Methods: A retrospective analysis including 91 NET patients (M47/F44; age 66 years, range 34-90 years) who completed four cycles of standard 177Lu-DOTATATE was conducted. SSTR-avid tumors were segmented from pretherapy SSTR-PET images using a semiautomatic workflow with the tumors labeled based on the anatomical regions. Multiple image-based features including total and organ-specific tumor volume and SSTR density along with clinicopathological biomarkers including Ki-67, chromogranin A (CgA) and alkaline phosphatase (ALP) were analyzed with respect to the PRRT response. Results: The median OS was 39.4 months (95% CI: 33.1-NA months), while the median PFS was 23.9 months (95% CI: 19.3-32.4 months). Total SSTR-avid tumor volume (HR = 3.6; P = 0.07) and bone tumor volume (HR = 1.5; P = 0.003) were associated with shorter OS. Also, total tumor volume (HR = 4.3; P = 0.01), liver tumor volume (HR = 1.8; P = 0.05) and bone tumor volume (HR = 1.4; P = 0.01) were associated with shorter PFS. Furthermore, the presence of large lesion volume with low SSTR uptake was correlated with worse OS (HR = 1.4; P = 0.03) and PFS (HR = 1.5; P = 0.003). Among the biomarkers, elevated baseline CgA and ALP showed a negative association with both OS (CgA: HR = 4.9; P = 0.003, ALP: HR = 52.6; P = 0.004) and PFS (CgA: HR = 4.2; P = 0.002, ALP: HR = 9.4; P = 0.06). Similarly, number of prior systemic treatments was associated with shorter OS (HR = 1.4; P = 0.003) and PFS (HR = 1.2; P = 0.05). Additionally, tumors originating from the midgut primary site demonstrated longer PFS, compared to the pancreas (HR = 1.6; P = 0.16), and those categorized as unknown primary (HR = 3.0; P = 0.002). Conclusion: Image-based features such as SSTR-avid tumor volume, bone tumor involvement, and the presence of large tumors with low SSTR expression demonstrated significant predictive value for PFS, suggesting potential clinical utility in NETs management. Moreover, elevated CgA and ALP, along with an increased number of prior systemic treatments, emerged as significant factors associated with worse PRRT outcomes.


Assuntos
Biomarcadores Tumorais , Tumores Neuroendócrinos , Octreotida , Compostos Organometálicos , Humanos , Tumores Neuroendócrinos/radioterapia , Tumores Neuroendócrinos/diagnóstico por imagem , Tumores Neuroendócrinos/patologia , Tumores Neuroendócrinos/metabolismo , Idoso , Pessoa de Meia-Idade , Compostos Organometálicos/uso terapêutico , Masculino , Feminino , Octreotida/análogos & derivados , Octreotida/uso terapêutico , Adulto , Estudos Retrospectivos , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Receptores de Somatostatina/metabolismo , Compostos Radiofarmacêuticos , Resultado do Tratamento , Cromogranina A/metabolismo , Fosfatase Alcalina/metabolismo , Antígeno Ki-67/metabolismo , Intervalo Livre de Progressão , Carga Tumoral
4.
EJNMMI Phys ; 11(1): 66, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39028439

RESUMO

BACKGROUND: Low-dose ungated CT is commonly used for total-body PET attenuation and scatter correction (ASC). However, CT-based ASC (CT-ASC) is limited by radiation dose risks of CT examinations, propagation of CT-based artifacts and potential mismatches between PET and CT. We demonstrate the feasibility of direct ASC for multi-tracer total-body PET in the image domain. METHODS: Clinical uEXPLORER total-body PET/CT datasets of [18F]FDG (N = 52), [18F]FAPI (N = 46) and [68Ga]FAPI (N = 60) were retrospectively enrolled in this study. We developed an improved 3D conditional generative adversarial network (cGAN) to directly estimate attenuation and scatter-corrected PET images from non-attenuation and scatter-corrected (NASC) PET images. The feasibility of the proposed 3D cGAN-based ASC was validated using four training strategies: (1) Paired 3D NASC and CT-ASC PET images from three tracers were pooled into one centralized server (CZ-ASC). (2) Paired 3D NASC and CT-ASC PET images from each tracer were individually used (DL-ASC). (3) Paired NASC and CT-ASC PET images from one tracer ([18F]FDG) were used to train the networks, while the other two tracers were used for testing without fine-tuning (NFT-ASC). (4) The pre-trained networks of (3) were fine-tuned with two other tracers individually (FT-ASC). We trained all networks in fivefold cross-validation. The performance of all ASC methods was evaluated by qualitative and quantitative metrics using CT-ASC as the reference. RESULTS: CZ-ASC, DL-ASC and FT-ASC showed comparable visual quality with CT-ASC for all tracers. CZ-ASC and DL-ASC resulted in a normalized mean absolute error (NMAE) of 8.51 ± 7.32% versus 7.36 ± 6.77% (p < 0.05), outperforming NASC (p < 0.0001) in [18F]FDG dataset. CZ-ASC, FT-ASC and DL-ASC led to NMAE of 6.44 ± 7.02%, 6.55 ± 5.89%, and 7.25 ± 6.33% in [18F]FAPI dataset, and NMAE of 5.53 ± 3.99%, 5.60 ± 4.02%, and 5.68 ± 4.12% in [68Ga]FAPI dataset, respectively. CZ-ASC, FT-ASC and DL-ASC were superior to NASC (p < 0.0001) and NFT-ASC (p < 0.0001) in terms of NMAE results. CONCLUSIONS: CZ-ASC, DL-ASC and FT-ASC demonstrated the feasibility of providing accurate and robust ASC for multi-tracer total-body PET, thereby reducing the radiation hazards to patients from redundant CT examinations. CZ-ASC and FT-ASC could outperform DL-ASC for cross-tracer total-body PET AC.

5.
J Imaging Inform Med ; 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38858260

RESUMO

To develop a robust segmentation model, encoding the underlying features/structures of the input data is essential to discriminate the target structure from the background. To enrich the extracted feature maps, contrastive learning and self-learning techniques are employed, particularly when the size of the training dataset is limited. In this work, we set out to investigate the impact of contrastive learning and self-learning on the performance of the deep learning-based semantic segmentation. To this end, three different datasets were employed used for brain tumor and hippocampus delineation from MR images (BraTS and Decathlon datasets, respectively) and kidney segmentation from CT images (Decathlon dataset). Since data augmentation techniques are also aimed at enhancing the performance of deep learning methods, a deformable data augmentation technique was proposed and compared with contrastive learning and self-learning frameworks. The segmentation accuracy for the three datasets was assessed with and without applying data augmentation, contrastive learning, and self-learning to individually investigate the impact of these techniques. The self-learning and deformable data augmentation techniques exhibited comparable performance with Dice indices of 0.913 ± 0.030 and 0.920 ± 0.022 for kidney segmentation, 0.890 ± 0.035 and 0.898 ± 0.027 for hippocampus segmentation, and 0.891 ± 0.045 and 0.897 ± 0.040 for lesion segmentation, respectively. These two approaches significantly outperformed the contrastive learning and the original model with Dice indices of 0.871 ± 0.039 and 0.868 ± 0.042 for kidney segmentation, 0.872 ± 0.045 and 0.865 ± 0.048 for hippocampus segmentation, and 0.870 ± 0.049 and 0.860 ± 0.058 for lesion segmentation, respectively. The combination of self-learning with deformable data augmentation led to a robust segmentation model with no outliers in the outcomes. This work demonstrated the beneficial impact of self-learning and deformable data augmentation on organ and lesion segmentation, where no additional training datasets are needed.

6.
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.

7.
J Appl Clin Med Phys ; 25(7): e14389, 2024 Jul.
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.


Assuntos
Método de Monte Carlo , Órgãos em Risco , Imagens de Fantasmas , Doses de Radiação , Radiografia Torácica , Tomografia Computadorizada por Raios X , Humanos , Tomografia Computadorizada por Raios X/métodos , Masculino , Feminino , Adulto , Órgãos em Risco/efeitos da radiação , Radiografia Torácica/métodos , Radiometria/métodos , Tórax/diagnóstico por imagem , Tórax/efeitos da radiação , Processamento de Imagem Assistida por Computador/métodos , Planejamento da Radioterapia Assistida por Computador/métodos
8.
BJR Open ; 6(1): tzae009, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38798693
9.
Phys Med Biol ; 69(11)2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38744305

RESUMO

This review casts a spotlight on intraoperative positron emission tomography (PET) scanners and the distinctive challenges they confront. Specifically, these systems contend with the necessity of partial coverage geometry, essential for ensuring adequate access to the patient. This inherently leans them towards limited-angle PET imaging, bringing along its array of reconstruction and geometrical sensitivity challenges. Compounding this, the need for real-time imaging in navigation systems mandates rapid acquisition and reconstruction times. For these systems, the emphasis is on dependable PET image reconstruction (without significant artefacts) while rapid processing takes precedence over the spatial resolution of the system. In contrast, specimen PET imagers are unburdened by the geometrical sensitivity challenges, thanks to their ability to leverage full coverage PET imaging geometries. For these devices, the focus shifts: high spatial resolution imaging takes precedence over rapid image reconstruction. This review concurrently probes into the technical complexities of both intraoperative and specimen PET imaging, shedding light on their recent designs, inherent challenges, and technological advancements.


Assuntos
Processamento de Imagem Assistida por Computador , Salas Cirúrgicas , Tomografia por Emissão de Pósitrons , Tomografia por Emissão de Pósitrons/instrumentação , Humanos , Processamento de Imagem Assistida por Computador/métodos
10.
Clin Genitourin Cancer ; 22(3): 102076, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38593599

RESUMO

The objective of this work was to review comparisons of the efficacy of 68Ga-PSMA-11 (prostate-specific membrane antigen) PET/CT and multiparametric magnetic resonance imaging (mpMRI) in the detection of prostate cancer among patients undergoing initial staging prior to radical prostatectomy or experiencing recurrent prostate cancer, based on histopathological data. A comprehensive search was conducted in PubMed and Web of Science, and relevant articles were analyzed with various parameters, including year of publication, study design, patient count, age, PSA (prostate-specific antigen) value, Gleason score, standardized uptake value (SUVmax), detection rate, treatment history, sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and PI-RADS (prostate imaging reporting and data system) scores. Only studies directly comparing PSMA-PET and mpMRI were considered, while those examining combined accuracy or focusing on either modality alone were excluded. In total, 24 studies comprising 1717 patients were analyzed, with the most common indication for screening being staging, followed by relapse. The findings indicated that 68Ga-PSMA-PET/CT effectively diagnosed prostate cancer in patients with suspected or confirmed disease, and both methods exhibited comparable efficacy in identifying lesion-specific information. However, notable heterogeneity was observed, highlighting the necessity for standardization of imaging and histopathology systems to mitigate inter-study variability. Future research should prioritize evaluating the combined diagnostic performance of both modalities to enhance sensitivity and reduce unnecessary biopsies. Overall, the utilization of PSMA-PET and mpMRI in combination holds substantial potential for significantly advancing the diagnosis and management of prostate cancer.


Assuntos
Isótopos de Gálio , Radioisótopos de Gálio , Imageamento por Ressonância Magnética Multiparamétrica , Recidiva Local de Neoplasia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Neoplasias da Próstata , Humanos , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/patologia , Neoplasias da Próstata/metabolismo , Masculino , Recidiva Local de Neoplasia/diagnóstico por imagem , Recidiva Local de Neoplasia/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Imageamento por Ressonância Magnética Multiparamétrica/métodos , Ácido Edético/análogos & derivados , Oligopeptídeos , Compostos Radiofarmacêuticos , Antígeno Prostático Específico/sangue , Antígeno Prostático Específico/metabolismo , Prostatectomia , Estadiamento de Neoplasias
11.
Ann Nucl Med ; 38(7): 493-507, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38575814

RESUMO

PURPOSE: This study aimed to examine the robustness of positron emission tomography (PET) radiomic features extracted via different segmentation methods before and after ComBat harmonization in patients with non-small cell lung cancer (NSCLC). METHODS: We included 120 patients (positive recurrence = 46 and negative recurrence = 74) referred for PET scanning as a routine part of their care. All patients had a biopsy-proven NSCLC. Nine segmentation methods were applied to each image, including manual delineation, K-means (KM), watershed, fuzzy-C-mean, region-growing, local active contour (LAC), and iterative thresholding (IT) with 40, 45, and 50% thresholds. Diverse image discretizations, both without a filter and with different wavelet decompositions, were applied to PET images. Overall, 6741 radiomic features were extracted from each image (749 radiomic features from each segmented area). Non-parametric empirical Bayes (NPEB) ComBat harmonization was used to harmonize the features. Linear Support Vector Classifier (LinearSVC) with L1 regularization For feature selection and Support Vector Machine classifier (SVM) with fivefold nested cross-validation was performed using StratifiedKFold with 'n_splits' set to 5 to predict recurrence in NSCLC patients and assess the impact of ComBat harmonization on the outcome. RESULTS: From 749 extracted radiomic features, 206 (27%) and 389 (51%) features showed excellent reliability (ICC ≥ 0.90) against segmentation method variation before and after NPEB ComBat harmonization, respectively. Among all, 39 features demonstrated poor reliability, which declined to 10 after ComBat harmonization. The 64 fixed bin widths (without any filter) and wavelets (LLL)-based radiomic features set achieved the best performance in terms of robustness against diverse segmentation techniques before and after ComBat harmonization. The first-order and GLRLM and also first-order and NGTDM feature families showed the largest number of robust features before and after ComBat harmonization, respectively. In terms of predicting recurrence in NSCLC, our findings indicate that using ComBat harmonization can significantly enhance machine learning outcomes, particularly improving the accuracy of watershed segmentation, which initially had fewer reliable features than manual contouring. Following the application of ComBat harmonization, the majority of cases saw substantial increase in sensitivity and specificity. CONCLUSION: Radiomic features are vulnerable to different segmentation methods. ComBat harmonization might be considered a solution to overcome the poor reliability of radiomic features.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Processamento de Imagem Assistida por Computador , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Neoplasias Pulmonares/diagnóstico por imagem , Processamento de Imagem Assistida por Computador/métodos , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Tomografia por Emissão de Pósitrons/métodos , Máquina de Vetores de Suporte , Adulto , Radiômica
12.
Phys Med ; 121: 103357, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38640631

RESUMO

PURPOSE: Large scintillation crystals-based gamma cameras play a crucial role in nuclear medicine imaging. In this study, a large field-of-view (FOV) gamma detector consisting of 48 square PMTs developed using a new readout electronics, reducing 48 (6 × 8) analog signals to 14 (6 + 8) analog sums of each row and column, with reduced complexity and cost while preserving image quality. METHODS: All 14 analog signals were converted to digital signals using AD9257 high-speed analog to digital (ADC) converters driven by the SPARTAN-6 family of field-programmable gate arrays (FPGA) in order to calculate the signal integrals. The positioning algorithm was based on the digital correlated signal enhancement (CSE) algorithm implemented in the acquisition software. The performance characteristics of the developed gamma camera were measured using the NEMA NU 1-2018 standards. RESULTS: The measured energy resolution of the developed detector was 8.7 % at 140 keV, with an intrinsic spatial resolution of 3.9 mm. The uniformity was within 0.6 %, while the linearity was within 0.1 %. CONCLUSION: The performance evaluation demonstrated that the developed detector has suitable specifications for high-end nuclear medicine imaging.


Assuntos
Câmaras gama , Eletrônica/instrumentação , Desenho de Equipamento , Algoritmos , Processamento de Imagem Assistida por Computador , Custos e Análise de Custo
13.
Med Phys ; 51(6): 4095-4104, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38629779

RESUMO

BACKGROUND: Contrast-enhanced computed tomography (CECT) provides much more information compared to non-enhanced CT images, especially for the differentiation of malignancies, such as liver carcinomas. Contrast media injection phase information is usually missing on public datasets and not standardized in the clinic even in the same region and language. This is a barrier to effective use of available CECT images in clinical research. PURPOSE: The aim of this study is to detect contrast media injection phase from CT images by means of organ segmentation and machine learning algorithms. METHODS: A total number of 2509 CT images split into four subsets of non-contrast (class #0), arterial (class #1), venous (class #2), and delayed (class #3) after contrast media injection were collected from two CT scanners. Seven organs including the liver, spleen, heart, kidneys, lungs, urinary bladder, and aorta along with body contour masks were generated by pre-trained deep learning algorithms. Subsequently, five first-order statistical features including average, standard deviation, 10, 50, and 90 percentiles extracted from the above-mentioned masks were fed to machine learning models after feature selection and reduction to classify the CT images in one of four above mentioned classes. A 10-fold data split strategy was followed. The performance of our methodology was evaluated in terms of classification accuracy metrics. RESULTS: The best performance was achieved by Boruta feature selection and RF model with average area under the curve of more than 0.999 and accuracy of 0.9936 averaged over four classes and 10 folds. Boruta feature selection selected all predictor features. The lowest classification was observed for class #2 (0.9888), which is already an excellent result. In the 10-fold strategy, only 33 cases from 2509 cases (∼1.4%) were misclassified. The performance over all folds was consistent. CONCLUSIONS: We developed a fast, accurate, reliable, and explainable methodology to classify contrast media phases which may be useful in data curation and annotation in big online datasets or local datasets with non-standard or no series description. Our model containing two steps of deep learning and machine learning may help to exploit available datasets more effectively.


Assuntos
Automação , Meios de Contraste , Processamento de Imagem Assistida por Computador , Aprendizado de Máquina , Tomografia Computadorizada por Raios X , Humanos , Processamento de Imagem Assistida por Computador/métodos , Radiografia Abdominal , Abdome/diagnóstico por imagem
14.
Med Biol Eng Comput ; 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38536580

RESUMO

This study investigated the impact of ComBat harmonization on the reproducibility of radiomic features extracted from magnetic resonance images (MRI) acquired on different scanners, using various data acquisition parameters and multiple image pre-processing techniques using a dedicated MRI phantom. Four scanners were used to acquire an MRI of a nonanatomic phantom as part of the TCIA RIDER database. In fast spin-echo inversion recovery (IR) sequences, several inversion durations were employed, including 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, and 3000 ms. In addition, a 3D fast spoiled gradient recalled echo (FSPGR) sequence was used to investigate several flip angles (FA): 2, 5, 10, 15, 20, 25, and 30 degrees. Nineteen phantom compartments were manually segmented. Different approaches were used to pre-process each image: Bin discretization, Wavelet filter, Laplacian of Gaussian, logarithm, square, square root, and gradient. Overall, 92 first-, second-, and higher-order statistical radiomic features were extracted. ComBat harmonization was also applied to the extracted radiomic features. Finally, the Intraclass Correlation Coefficient (ICC) and Kruskal-Wallis's (KW) tests were implemented to assess the robustness of radiomic features. The number of non-significant features in the KW test ranged between 0-5 and 29-74 for various scanners, 31-91 and 37-92 for three times tests, 0-33 to 34-90 for FAs, and 3-68 to 65-89 for IRs before and after ComBat harmonization, with different image pre-processing techniques, respectively. The number of features with ICC over 90% ranged between 0-8 and 6-60 for various scanners, 11-75 and 17-80 for three times tests, 3-83 to 9-84 for FAs, and 3-49 to 3-63 for IRs before and after ComBat harmonization, with different image pre-processing techniques, respectively. The use of various scanners, IRs, and FAs has a great impact on radiomic features. However, the majority of scanner-robust features is also robust to IR and FA. Among the effective parameters in MR images, several tests in one scanner have a negligible impact on radiomic features. Different scanners and acquisition parameters using various image pre-processing might affect radiomic features to a large extent. ComBat harmonization might significantly impact the reproducibility of MRI radiomic features.

15.
Phys Eng Sci Med ; 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38512435

RESUMO

Manual segmentation poses a time-consuming challenge for disease quantification, therapy evaluation, treatment planning, and outcome prediction. Convolutional neural networks (CNNs) hold promise in accurately identifying tumor locations and boundaries in PET scans. However, a major hurdle is the extensive amount of supervised and annotated data necessary for training. To overcome this limitation, this study explores semi-supervised approaches utilizing unlabeled data, specifically focusing on PET images of diffuse large B-cell lymphoma (DLBCL) and primary mediastinal large B-cell lymphoma (PMBCL) obtained from two centers. We considered 2-[18F]FDG PET images of 292 patients PMBCL (n = 104) and DLBCL (n = 188) (n = 232 for training and validation, and n = 60 for external testing). We harnessed classical wisdom embedded in traditional segmentation methods, such as the fuzzy clustering loss function (FCM), to tailor the training strategy for a 3D U-Net model, incorporating both supervised and unsupervised learning approaches. Various supervision levels were explored, including fully supervised methods with labeled FCM and unified focal/Dice loss, unsupervised methods with robust FCM (RFCM) and Mumford-Shah (MS) loss, and semi-supervised methods combining FCM with supervised Dice loss (MS + Dice) or labeled FCM (RFCM + FCM). The unified loss function yielded higher Dice scores (0.73 ± 0.11; 95% CI 0.67-0.8) than Dice loss (p value < 0.01). Among the semi-supervised approaches, RFCM + αFCM (α = 0.3) showed the best performance, with Dice score of 0.68 ± 0.10 (95% CI 0.45-0.77), outperforming MS + αDice for any supervision level (any α) (p < 0.01). Another semi-supervised approach with MS + αDice (α = 0.2) achieved Dice score of 0.59 ± 0.09 (95% CI 0.44-0.76) surpassing other supervision levels (p < 0.01). Given the time-consuming nature of manual delineations and the inconsistencies they may introduce, semi-supervised approaches hold promise for automating medical imaging segmentation workflows.

16.
Med Phys ; 51(7): 4736-4747, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38335175

RESUMO

BACKGROUND: Notwithstanding the encouraging results of previous studies reporting on the efficiency of deep learning (DL) in COVID-19 prognostication, clinical adoption of the developed methodology still needs to be improved. To overcome this limitation, we set out to predict the prognosis of a large multi-institutional cohort of patients with COVID-19 using a DL-based model. PURPOSE: This study aimed to evaluate the performance of deep privacy-preserving federated learning (DPFL) in predicting COVID-19 outcomes using chest CT images. METHODS: After applying inclusion and exclusion criteria, 3055 patients from 19 centers, including 1599 alive and 1456 deceased, were enrolled in this study. Data from all centers were split (randomly with stratification respective to each center and class) into a training/validation set (70%/10%) and a hold-out test set (20%). For the DL model, feature extraction was performed on 2D slices, and averaging was performed at the final layer to construct a 3D model for each scan. The DensNet model was used for feature extraction. The model was developed using centralized and FL approaches. For FL, we employed DPFL approaches. Membership inference attack was also evaluated in the FL strategy. For model evaluation, different metrics were reported in the hold-out test sets. In addition, models trained in two scenarios, centralized and FL, were compared using the DeLong test for statistical differences. RESULTS: The centralized model achieved an accuracy of 0.76, while the DPFL model had an accuracy of 0.75. Both the centralized and DPFL models achieved a specificity of 0.77. The centralized model achieved a sensitivity of 0.74, while the DPFL model had a sensitivity of 0.73. A mean AUC of 0.82 and 0.81 with 95% confidence intervals of (95% CI: 0.79-0.85) and (95% CI: 0.77-0.84) were achieved by the centralized model and the DPFL model, respectively. The DeLong test did not prove statistically significant differences between the two models (p-value = 0.98). The AUC values for the inference attacks fluctuate between 0.49 and 0.51, with an average of 0.50 ± 0.003 and 95% CI for the mean AUC of 0.500 to 0.501. CONCLUSION: The performance of the proposed model was comparable to centralized models while operating on large and heterogeneous multi-institutional datasets. In addition, the model was resistant to inference attacks, ensuring the privacy of shared data during the training process.


Assuntos
COVID-19 , Aprendizado Profundo , Tomografia Computadorizada por Raios X , COVID-19/diagnóstico por imagem , Humanos , Prognóstico , Masculino , Feminino , Idoso , Pessoa de Meia-Idade , Privacidade , Radiografia Torácica , Conjuntos de Dados como Assunto
17.
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
18.
Z Med Phys ; 2024 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-38302292

RESUMO

In positron emission tomography (PET), attenuation and scatter corrections are necessary steps toward accurate quantitative reconstruction of the radiopharmaceutical distribution. Inspired by recent advances in deep learning, many algorithms based on convolutional neural networks have been proposed for automatic attenuation and scatter correction, enabling applications to CT-less or MR-less PET scanners to improve performance in the presence of CT-related artifacts. A known characteristic of PET imaging is to have varying tracer uptakes for various patients and/or anatomical regions. However, existing deep learning-based algorithms utilize a fixed model across different subjects and/or anatomical regions during inference, which could result in spurious outputs. In this work, we present a novel deep learning-based framework for the direct reconstruction of attenuation and scatter-corrected PET from non-attenuation-corrected images in the absence of structural information in the inference. To deal with inter-subject and intra-subject uptake variations in PET imaging, we propose a novel model to perform subject- and region-specific filtering through modulating the convolution kernels in accordance to the contextual coherency within the neighboring slices. This way, the context-aware convolution can guide the composition of intermediate features in favor of regressing input-conditioned and/or region-specific tracer uptakes. We also utilized a large cohort of 910 whole-body studies for training and evaluation purposes, which is more than one order of magnitude larger than previous works. In our experimental studies, qualitative assessments showed that our proposed CT-free method is capable of producing corrected PET images that accurately resemble ground truth images corrected with the aid of CT scans. For quantitative assessments, we evaluated our proposed method over 112 held-out subjects and achieved an absolute relative error of 14.30±3.88% and a relative error of -2.11%±2.73% in whole-body.

19.
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
20.
Radiat Oncol ; 19(1): 12, 2024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-38254203

RESUMO

BACKGROUND: This study aimed to investigate the value of clinical, radiomic features extracted from gross tumor volumes (GTVs) delineated on CT images, dose distributions (Dosiomics), and fusion of CT and dose distributions to predict outcomes in head and neck cancer (HNC) patients. METHODS: A cohort of 240 HNC patients from five different centers was obtained from The Cancer Imaging Archive. Seven strategies, including four non-fusion (Clinical, CT, Dose, DualCT-Dose), and three fusion algorithms (latent low-rank representation referred (LLRR),Wavelet, weighted least square (WLS)) were applied. The fusion algorithms were used to fuse the pre-treatment CT images and 3-dimensional dose maps. Overall, 215 radiomics and Dosiomics features were extracted from the GTVs, alongside with seven clinical features incorporated. Five feature selection (FS) methods in combination with six machine learning (ML) models were implemented. The performance of the models was quantified using the concordance index (CI) in one-center-leave-out 5-fold cross-validation for overall survival (OS) prediction considering the time-to-event. RESULTS: The mean CI and Kaplan-Meier curves were used for further comparisons. The CoxBoost ML model using the Minimal Depth (MD) FS method and the glmnet model using the Variable hunting (VH) FS method showed the best performance with CI = 0.73 ± 0.15 for features extracted from LLRR fused images. In addition, both glmnet-Cindex and Coxph-Cindex classifiers achieved a CI of 0.72 ± 0.14 by employing the dose images (+ incorporated clinical features) only. CONCLUSION: Our results demonstrated that clinical features, Dosiomics and fusion of dose and CT images by specific ML-FS models could predict the overall survival of HNC patients with acceptable accuracy. Besides, the performance of ML methods among the three different strategies was almost comparable.


Assuntos
Neoplasias de Cabeça e Pescoço , Radiômica , Humanos , Prognóstico , Neoplasias de Cabeça e Pescoço/diagnóstico por imagem , Neoplasias de Cabeça e Pescoço/radioterapia , Aprendizado de Máquina , Tomografia Computadorizada por Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA