Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 222
1.
Phys Med ; 121: 103357, 2024 May.
Article En | MEDLINE | ID: mdl-38640631

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.


Gamma Cameras , Electronics/instrumentation , Equipment Design , Algorithms , Image Processing, Computer-Assisted , Costs and Cost Analysis
2.
Phys Med ; 121: 103366, 2024 May.
Article En | MEDLINE | ID: mdl-38657425

The purpose of this investigation is to quantify the spatial heterogeneity of prostate-specific membrane antigen (PSMA) positron emission tomography (PET) uptake within parotid glands. We aim to quantify patterns in well-defined regions to facilitate further investigations. Furthermore, we investigate whether uptake is correlated with computed tomography (CT) texture features. METHODS: Parotid glands from [18F]DCFPyL PSMA PET/CT images of 30 prostate cancer patients were analyzed. Uptake patterns were assessed with various segmentation schemes. Spearman's rank correlation coefficient was calculated between PSMA PET uptake and feature values of a Grey Level Run Length Matrix using a long and short run length emphasis (GLRLML and GLRLMS) in subregions of the parotid gland. RESULTS: PSMA PET uptake was significantly higher (p < 0.001) in lateral/posterior regions of the glands than anterior/medial regions. Maximum uptake was found in the lateral half of parotid glands in 50 out of 60 glands. The difference in SUVmean between parotid halves is greatest when parotids are divided by a plane separating the anterior/medial and posterior/lateral halves symmetrically (out of 120 bisections tested). PSMA PET uptake was significantly correlated with CT GLRLML (p < 0.001), and anti-correlated with CT GLRLMS (p < 0.001). CONCLUSION: Uptake of PSMA PET is heterogeneous within parotid glands, with uptake biased towards lateral/posterior regions. Uptake within parotid glands was strongly correlated with CT texture feature maps.


Glutamate Carboxypeptidase II , Lysine/analogs & derivatives , Parotid Gland , Positron Emission Tomography Computed Tomography , Urea/analogs & derivatives , Humans , Parotid Gland/diagnostic imaging , Parotid Gland/metabolism , Glutamate Carboxypeptidase II/metabolism , Male , Ligands , Antigens, Surface/metabolism , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/metabolism , Biological Transport , Aged , Middle Aged
3.
Front Oncol ; 14: 1320371, 2024.
Article En | MEDLINE | ID: mdl-38559559

Introduction: Computational models yield valuable insights into biological interactions not fully elucidated by experimental approaches. This study investigates an innovative spatiotemporal model for simulating the controlled release and dispersion of radiopharmaceutical therapy (RPT) using 177Lu-PSMA, a prostate-specific membrane antigen (PSMA) targeted radiopharmaceutical, within solid tumors via a dual-release implantable delivery system. Local delivery of anticancer agents presents a strategic approach to mitigate adverse effects while optimizing therapeutic outcomes. Methods: This study evaluates various factors impacting RPT efficacy, including hypoxia region extension, binding affinity, and initial drug dosage, employing a novel 3-dimensional computational model. Analysis gauges the influence of these factors on radiopharmaceutical agent concentration within the tumor microenvironment. Furthermore, spatial and temporal radiopharmaceutical distribution within both the tumor and surrounding tissue is explored. Results: Analysis indicates a significantly higher total concentration area under the curve within the tumor region compared to surrounding normal tissue. Moreover, drug distribution exhibits notably superior efficacy compared to the radiation source. Additionally, low microvascular density in extended hypoxia regions enhances drug availability, facilitating improved binding to PSMA receptors and enhancing therapeutic effectiveness. Reductions in the dissociation constant (KD) lead to heightened binding affinity and increased internalized drug concentration. Evaluation of initial radioactivities (7.1×107, 7.1×108, and 7.1×109 [Bq]) indicates that an activity of 7.1×108 [Bq] offers a favorable balance between tumor cell elimination and minimal impact on normal tissues. Discussion: These findings underscore the potential of localized radiopharmaceutical delivery strategies and emphasize the crucial role of released drugs relative to the radiation source (implant) in effective tumor treatment. Decreasing the proximity of the drug to the microvascular network and enhancing its distribution within the tumor promote a more effective therapeutic outcome. The study furnishes valuable insights for future experimental investigations and clinical trials, aiming to refine medication protocols and minimize reliance on in vivo testing.

4.
NPJ Syst Biol Appl ; 10(1): 39, 2024 Apr 12.
Article En | MEDLINE | ID: mdl-38609421

Lutetium-177 prostate-specific membrane antigen (177Lu-PSMA)-targeted radiopharmaceutical therapy is a clinically approved treatment for patients with metastatic castration-resistant prostate cancer (mCRPC). Even though common practice reluctantly follows "one size fits all" approach, medical community believes there is significant room for deeper understanding and personalization of radiopharmaceutical therapies. To pursue this aim, we present a 3-dimensional spatiotemporal radiopharmaceutical delivery model based on clinical imaging data to simulate pharmacokinetic of 177Lu-PSMA within the prostate tumors. The model includes interstitial flow, radiopharmaceutical transport in tissues, receptor cycles, association/dissociation with ligands, synthesis of PSMA receptors, receptor recycling, internalization of radiopharmaceuticals, and degradation of receptors and drugs. The model was studied for a range of values for injection amount (100-1000 nmol), receptor density (10-500 nmol•l-1), and recycling rate of receptors (10-4 to 10-1 min-1). Furthermore, injection type, different convection-diffusion-reaction mechanisms, characteristic time scales, and length scales are discussed. The study found that increasing receptor density, ligand amount, and labeled ligands improved radiopharmaceutical uptake in the tumor. A high receptor recycling rate (0.1 min-1) increased radiopharmaceutical concentration by promoting repeated binding to tumor cell receptors. Continuous infusion results in higher radiopharmaceutical concentrations within tumors compared to bolus administration. These insights are crucial for advancing targeted therapy for prostate cancer by understanding the mechanism of radiopharmaceutical distribution in tumors. Furthermore, measures of characteristic length and advection time scale were computed. The presented spatiotemporal tumor transport model can analyze different physiological parameters affecting 177Lu-PSMA delivery.


Prostatic Neoplasms , Radiopharmaceuticals , Male , Humans , Prostatic Neoplasms/radiotherapy , Biological Transport , Diffusion
5.
Phys Med ; 121: 103336, 2024 May.
Article En | MEDLINE | ID: mdl-38626637

PURPOSE: We aimed to investigate whether a clinically feasible dual time-point (DTP) approach can accurately estimate the metabolic uptake rate constant (Ki) and to explore reliable acquisition times through simulations and clinical assessment considering patient comfort and quantification accuracy. METHODS: We simulated uptake kinetics in different tumors for four sets of DTP PET images within the routine clinical static acquisition at 60-min post-injection (p.i.). We determined Ki for a total of 81 lesions. Ki quantification from full dynamic PET data (Patlak-Ki) and Ki from DTP (DTP-Ki) were compared. In addition, we scaled a population-based input function (PBIFscl) with the image-derived blood pool activity sampled at different time points to assess the best scaling time-point for Ki quantifications in the simulation data. RESULTS: In the simulation study, Ki estimated using DTP via (30,60-min), (30,90-min), (60,90-min), and (60,120-min) samples showed strong correlations (r ≥ 0.944, P < 0.0001) with the true value of Ki. The DTP results with the PBIFscl at 60-min time-point in (30,60-min), (60,90-min), and (60,120-min) were linearly related to the true Ki with a slope of 1.037, 1.008, 1.013 and intercept of -6 × 10-4, 2 × 10-5, 5 × 10-5, respectively. In a clinical study, strong correlations (r ≥ 0.833, P < 0.0001) were observed between Patlak-Ki and DTP-Ki. The Patlak-derived mean values of Ki, tumor-to-background-ratio, signal-to-noise-ratio, and contrast-to-noise-ratio were linearly correlated with the DTP method. CONCLUSIONS: Besides calculating the retention index as a commonly used quantification parameter inDTP imaging,our DTP method can accurately estimate Ki.


Feasibility Studies , Fluorodeoxyglucose F18 , Positron-Emission Tomography , Humans , Fluorodeoxyglucose F18/metabolism , Positron-Emission Tomography/methods , Time Factors , Image Processing, Computer-Assisted/methods , Kinetics , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Biological Transport , Male , Female , Middle Aged , Aged , Computer Simulation
6.
Cancers (Basel) ; 16(6)2024 Mar 08.
Article En | MEDLINE | ID: mdl-38539425

OBJECTIVES: Accurate outcome prediction is important for making informed clinical decisions in cancer treatment. In this study, we assessed the feasibility of using changes in radiomic features over time (Delta radiomics: absolute and relative) following chemotherapy, to predict relapse/progression and time to progression (TTP) of primary mediastinal large B-cell lymphoma (PMBCL) patients. MATERIAL AND METHODS: Given the lack of standard staging PET scans until 2011, only 31 out of 103 PMBCL patients in our retrospective study had both pre-treatment and end-of-treatment (EoT) scans. Consequently, our radiomics analysis focused on these 31 patients who underwent [18F]FDG PET-CT scans before and after R-CHOP chemotherapy. Expert manual lesion segmentation was conducted on their scans for delta radiomics analysis, along with an additional 19 EoT scans, totaling 50 segmented scans for single time point analysis. Radiomics features (on PET and CT), along with maximum and mean standardized uptake values (SUVmax and SUVmean), total metabolic tumor volume (TMTV), tumor dissemination (Dmax), total lesion glycolysis (TLG), and the area under the curve of cumulative standardized uptake value-volume histogram (AUC-CSH) were calculated. We additionally applied longitudinal analysis using radial mean intensity (RIM) changes. For prediction of relapse/progression, we utilized the individual coefficient approximation for risk estimation (ICARE) and machine learning (ML) techniques (K-Nearest Neighbor (KNN), Linear Discriminant Analysis (LDA), and Random Forest (RF)) including sequential feature selection (SFS) following correlation analysis for feature selection. For TTP, ICARE and CoxNet approaches were utilized. In all models, we used nested cross-validation (CV) (with 10 outer folds and 5 repetitions, along with 5 inner folds and 20 repetitions) after balancing the dataset using Synthetic Minority Oversampling TEchnique (SMOTE). RESULTS: To predict relapse/progression using Delta radiomics between the baseline (staging) and EoT scans, the best performances in terms of accuracy and F1 score (F1 score is the harmonic mean of precision and recall, where precision is the ratio of true positives to the sum of true positives and false positives, and recall is the ratio of true positives to the sum of true positives and false negatives) were achieved with ICARE (accuracy = 0.81 ± 0.15, F1 = 0.77 ± 0.18), RF (accuracy = 0.89 ± 0.04, F1 = 0.87 ± 0.04), and LDA (accuracy = 0.89 ± 0.03, F1 = 0.89 ± 0.03), that are higher compared to the predictive power achieved by using only EoT radiomics features. For the second category of our analysis, TTP prediction, the best performer was CoxNet (LASSO feature selection) with c-index = 0.67 ± 0.06 when using baseline + Delta features (inclusion of both baseline and Delta features). The TTP results via Delta radiomics were comparable to the use of radiomics features extracted from EoT scans for TTP analysis (c-index = 0.68 ± 0.09) using CoxNet (with SFS). The performance of Deauville Score (DS) for TTP was c-index = 0.66 ± 0.09 for n = 50 and 0.67 ± 03 for n = 31 cases when using EoT scans with no significant differences compared to the radiomics signature from either EoT scans or baseline + Delta features (p-value> 0.05). CONCLUSION: This work demonstrates the potential of Delta radiomics and the importance of using EoT scans to predict progression and TTP from PMBCL [18F]FDG PET-CT scans.

7.
Phys Eng Sci Med ; 2024 Mar 21.
Article En | MEDLINE | ID: mdl-38512435

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.

9.
Phys Med Biol ; 69(8)2024 Apr 09.
Article En | MEDLINE | ID: mdl-38513292

Objective. To simultaneously deblur and supersample prostate specific membrane antigen (PSMA) positron emission tomography (PET) images using neural blind deconvolution.Approach. Blind deconvolution is a method of estimating the hypothetical 'deblurred' image along with the blur kernel (related to the point spread function) simultaneously. Traditionalmaximum a posterioriblind deconvolution methods require stringent assumptions and suffer from convergence to a trivial solution. A method of modelling the deblurred image and kernel with independent neural networks, called 'neural blind deconvolution' had demonstrated success for deblurring 2D natural images in 2020. In this work, we adapt neural blind deconvolution to deblur PSMA PET images while simultaneous supersampling to double the original resolution. We compare this methodology with several interpolation methods in terms of resultant blind image quality metrics and test the model's ability to predict accurate kernels by re-running the model after applying artificial 'pseudokernels' to deblurred images. The methodology was tested on a retrospective set of 30 prostate patients as well as phantom images containing spherical lesions of various volumes.Main results. Neural blind deconvolution led to improvements in image quality over other interpolation methods in terms of blind image quality metrics, recovery coefficients, and visual assessment. Predicted kernels were similar between patients, and the model accurately predicted several artificially-applied pseudokernels. Localization of activity in phantom spheres was improved after deblurring, allowing small lesions to be more accurately defined.Significance. The intrinsically low spatial resolution of PSMA PET leads to partial volume effects (PVEs) which negatively impact uptake quantification in small regions. The proposed method can be used to mitigate this issue, and can be straightforwardly adapted for other imaging modalities.


Image Processing, Computer-Assisted , Positron-Emission Tomography , Male , Humans , Image Processing, Computer-Assisted/methods , Retrospective Studies , Positron-Emission Tomography/methods
10.
Article En | MEDLINE | ID: mdl-38326655

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.

11.
Z Med Phys ; 2024 Jan 31.
Article En | MEDLINE | ID: mdl-38302292

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.

12.
Radiology ; 310(2): e231319, 2024 Feb.
Article En | MEDLINE | ID: mdl-38319168

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.


Image Processing, Computer-Assisted , Radiomics , Humans , Reproducibility of Results , Biomarkers , Multimodal Imaging
13.
Eur J Radiol ; 172: 111349, 2024 Mar.
Article En | MEDLINE | ID: mdl-38310673

PURPOSE: Radiomics analysis of oncologic positron emission tomography (PET) images is an area of significant activity and potential. The reproducibility of radiomics features is an important consideration for routine clinical use. This preliminary study investigates the robustness of radiomics features in PSMA-PET images across penalized-likelihood (Q.Clear) and standard ordered subset expectation maximization (OSEM) reconstruction algorithms and their setting parameters in phantom and prostate cancer (PCa) patients. METHOD: A NEMA image quality (IQ) phantom and 8 PCa patients were selected for phantom and patient analyses, respectively. PET images were reconstructed using Q.Clear (reconstruction ß-value: 100-700, at intervals of 100 for both NEMA IQ phantom and patients) and OSEM (duration: 15sec, 30sec, 1 min, 2 min, 3 min, 4 min and 5 min for NEMA phantom and duration: 30 s, 1 min and 2 min for patients) reconstruction methods. Subsequently, 129 radiomic features were extracted from the reconstructed images. The coefficient of variation (COV) of each feature across reconstruction methods and their parameters was calculated to determine feature robustness. RESULTS: The extracted radiomics features showed a different range of variability, depending on the reconstruction algorithms and setting parameters. Specifically, 23.0 % and 53.5 % of features were found as robust against ß-value variations in Q.Clear and different durations in OSEM reconstruction algorithms, respectively. Taking into account the two algorithms and their parameters, eleven features (8.5 %) showed COV ≤ 5 % and eighteen (14 %) showed 5 % 20 %. The mean COVs of the extracted radiomics features were significantly different between the two reconstruction methods (p < 0.05) except for the phantom morphological features. CONCLUSIONS: All radiomics features were affected by reconstruction methods and parameters, but features with small or very small variations are considered better candidates for reproducible quantification of either tumor or metastatic tissues in clinical trials. There is a need for standardization before the implementation of PET radiomics in clinical practice.


Image Processing, Computer-Assisted , Radiomics , Male , Humans , Reproducibility of Results , Image Processing, Computer-Assisted/methods , Positron-Emission Tomography/methods , Algorithms , Phantoms, Imaging , Positron Emission Tomography Computed Tomography
14.
Med Phys ; 2024 Feb 13.
Article En | MEDLINE | ID: mdl-38348927

BACKGROUND: Phantoms are commonly used to evaluate and compare the performance of imaging systems given the known ground truth. Positron emission tomography (PET) scanners are routinely validated using the NEMA image quality phantom, in which lesions are modeled using 10 to 37 mm fillable spheres. The NEMA phantom neglects, however, to model focal (3-10-mm), high-uptake lesions that are increasingly observed in prostate-specific membrane antigen (PSMA) PET images. PSMA-targeting radiopharmaceuticals allow for enhanced detection of metastatic prostate cancers. As such, there is significant need to develop an updated phantom which considers both the quantitative and lesion detectability of this new paradigm in oncological PET imaging. PURPOSE: In this work, we present the Quantitative PET Prostate Phantom (Q3P); a portable and modular phantom that can be used to improve and harmonize imaging protocols for 18 F-PSMA PET scans. METHODS: A one-piece cylindrical phantom was designed effectively in two halves, which we call modules. Module 1 was designed to mimic lesions in the presence of background, and Module 2 mimicked very high contrast conditions (i.e., very low background) that can be observed in 18 F-PSMA PET scans. Shell-less radioactive spheres (3-16-mm) were cast using epoxy resin mixed with sodium-22 (22 Na), a long half-life positron emitter with positron range similar to 18 F. To establish realistic lesion contrast, the 22 Na spheres were mounted in a cylindrical chamber that can be filled with an 18 F background (module 1). Thirteen exchangeable spherical cavity inserts (3-37-mm) were machined in two parts and solvent welded together, and filled with 18 F (50 kBq/mL) to model lesions with very high contrast (module 2). Five 2.5-min PET scans were acquired on a 5-ring GE Discovery MI PET/CT scanner (General Electric, USA). Lesions were segmented using 41% of SUVmax fixed thresholding (41% FT) and recovery coefficients (RCs) were computed from 5 noise realizations. RESULTS: The manufactured phantom is portable (5.7 kg) and scan preparation takes less than 40 min. The total 22 Na activity is 250 kBq, allowing it to be shipped as an exempt package under International Atomic Energy Agency (IAEA) regulations. Recovery coefficients, computed using PSF modeling and no post-reconstruction smoothing, were 130.3% (16 mm), 147.1% (10 mm), 87.2% (6 mm), and 7.0% (3 mm) for RCmax , which decreased to 91.1% (16 mm), 90.6% (10 mm), 53.2% (6 mm), and 3.6% (3 mm) for RCmean in the 22 Na spheres. Comparatively, 18 F sphere recovery was 110.7% (17 mm), 123.6% (10 mm), 106.5% (7 mm), and 23.3% (3 mm) for RCmax , which was reduced to 76.7% (17 mm), 77.7% (10 mm), 66.8% (7 mm), and 13.5% (3 mm), for RCmean . CONCLUSIONS: A standardized imaging phantom was developed for lesion quantification assessment in 18 F-PSMA PET images. The phantom is configurable, providing users with the opportunity to modify background activity levels or sphere sizes according to clinical demands. Distributed to the community, the Q3P phantom has the potential to enable better assessment of lesion quantification and harmonization of 18 F-PSMA PET imaging, which may lead to more robust predictive metrics and better outcome prediction in metastatic prostate cancer.

15.
EJNMMI Radiopharm Chem ; 9(1): 6, 2024 Jan 22.
Article En | MEDLINE | ID: mdl-38252191

BACKGROUND: We aimed to develop a publicly shared computational physiologically based pharmacokinetic (PBPK) model to reliably simulate and analyze radiopharmaceutical therapies (RPTs), including probing of hot-cold ligand competitions as well as alternative injection scenarios and drug designs, towards optimal therapies. RESULTS: To handle the complexity of PBPK models (over 150 differential equations), a scalable modeling notation called the "reaction graph" is introduced, enabling easy inclusion of various interactions. We refer to this as physiologically based radiopharmacokinetic (PBRPK) modeling, fine-tuned specifically for radiopharmaceuticals. As three important applications, we used our PBRPK model to (1) study the effect of competition between hot and cold species on delivered doses to tumors and organs at risk. In addition, (2) we evaluated an alternative paradigm of utilizing multi-bolus injections in RPTs instead of prevalent single injections. Finally, (3) we used PBRPK modeling to study the impact of varying albumin-binding affinities by ligands, and the implications for RPTs. We found that competition between labeled and unlabeled ligands can lead to non-linear relations between injected activity and the delivered dose to a particular organ, in the sense that doubling the injected activity does not necessarily result in a doubled dose delivered to a particular organ (a false intuition from external beam radiotherapy). In addition, we observed that fractionating injections can lead to a higher payload of dose delivery to organs, though not a differential dose delivery to the tumor. By contrast, we found out that increased albumin-binding affinities of the injected ligands can lead to such a differential effect in delivering more doses to tumors, and this can be attributed to several factors that PBRPK modeling allows us to probe. CONCLUSIONS: Advanced computational PBRPK modeling enables simulation and analysis of a variety of intervention and drug design scenarios, towards more optimal delivery of RPTs.

16.
Eur J Nucl Med Mol Imaging ; 51(6): 1516-1529, 2024 May.
Article En | MEDLINE | ID: mdl-38267686

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.


Octreotide , Octreotide/analogs & derivatives , Organometallic Compounds , Radiometry , Radiopharmaceuticals , Single Photon Emission Computed Tomography Computed Tomography , Humans , Octreotide/therapeutic use , Organometallic Compounds/therapeutic use , Single Photon Emission Computed Tomography Computed Tomography/methods , Radiometry/methods , Radiopharmaceuticals/therapeutic use , Precision Medicine/methods , Deep Learning , Male , Female , Monte Carlo Method , Image Processing, Computer-Assisted/methods , Neuroendocrine Tumors/radiotherapy , Neuroendocrine Tumors/diagnostic imaging
17.
Diagnostics (Basel) ; 14(2)2024 Jan 14.
Article En | MEDLINE | ID: mdl-38248059

Radiotheranostics refers to the pairing of radioactive imaging biomarkers with radioactive therapeutic compounds that deliver ionizing radiation. Given the introduction of very promising radiopharmaceuticals, the radiotheranostics approach is creating a novel paradigm in personalized, targeted radionuclide therapies (TRTs), also known as radiopharmaceuticals (RPTs). Radiotherapeutic pairs targeting somatostatin receptors (SSTR) and prostate-specific membrane antigens (PSMA) are increasingly being used to diagnose and treat patients with metastatic neuroendocrine tumors (NETs) and prostate cancer. In parallel, radiomics and artificial intelligence (AI), as important areas in quantitative image analysis, are paving the way for significantly enhanced workflows in diagnostic and theranostic fields, from data and image processing to clinical decision support, improving patient selection, personalized treatment strategies, response prediction, and prognostication. Furthermore, AI has the potential for tremendous effectiveness in patient dosimetry which copes with complex and time-consuming tasks in the RPT workflow. The present work provides a comprehensive overview of radiomics and AI application in radiotheranostics, focusing on pairs of SSTR- or PSMA-targeting radioligands, describing the fundamental concepts and specific imaging/treatment features. Our review includes ligands radiolabeled by 68Ga, 18F, 177Lu, 64Cu, 90Y, and 225Ac. Specifically, contributions via radiomics and AI towards improved image acquisition, reconstruction, treatment response, segmentation, restaging, lesion classification, dose prediction, and estimation as well as ongoing developments and future directions are discussed.

18.
Biomed Phys Eng Express ; 10(2)2024 Feb 05.
Article En | MEDLINE | ID: mdl-38271732

Objective. Xerostomia and radiation-induced salivary gland dysfunction remain a common side effect for head-and-neck radiotherapy patients, and attempts have been made to quantify the heterogeneity of the dose response within parotid glands. Prostate Specific Membrane Antigen (PSMA) ligands have demonstrated high uptake in salivary glands, which has been shown to correlate with gland functionality. Here we compare several models of parotid gland subregional relative importance with PSMA positron emission tomography (PET) uptake. We then develop a predictive model for Clarket al's relative importance estimates using PSMA PET and CT radiomic features, and demonstrate a methodology for predicting patient-specific importance deviations from the population.Approach. Intra-parotid gland uptake was compared with four regional importance models using 30 [18F]DCFPyL PSMA PET images. The correlation of uptake and importance was ascertained when numerous non-overlapping subregions were defined, while a paired t-test was used to compare binary region pairs. A radiomics-based predictive model of population importance was developed using a double cross-validation methodology. A model was then devised for supplementing population-level subregional importance estimates for each patient using patient-specific radiomic features.Main Results. Anticorrelative relationships were found to exist between PSMA PET uptake and four independent models of subregional parotid gland importance from the literature. Kernel Ridge Regression with principal component analysis feature selection performed best over test sets (Mean Absolute Error = 0.08), with gray level co-occurrence matrix (GLCM) features being particularly important. Deblurring PSMA PET images with neural blind deconvolution strengthened correlations and improved model performance.Significance. This study suggests that regions of relatively low PSMA PET uptake in parotid glands may exhibit relatively high dose-sensitivity. We've demonstrated the utility of PSMA PET radiomic features for predicting relative importance within subregions of parotid glands. PSMA PET appears to be a promising quantitative imaging modality for analyzing salivary gland functionality.


Parotid Gland , Positron Emission Tomography Computed Tomography , Humans , Head , Parotid Gland/diagnostic imaging , Positron-Emission Tomography
19.
Comput Methods Programs Biomed ; 245: 108004, 2024 Mar.
Article En | MEDLINE | ID: mdl-38215660

BACKGROUND AND OBJECTIVE: 177Lu-labeled prostate-specific membrane antigen (PSMA) radiopharmaceutical therapy (RPT) represents a pivotal advancement in addressing prostate cancer. However, existing therapies, while promising, remain incompletely understood and optimized. Computational models offer potential insights into RPTs, aiding in clinical drug delivery enhancement. In this study, we investigate the impact of various physiological parameters on the delivery of 177Lu-PSMA-617 RPT using the convection-diffusion-reaction (CDR) model. METHODS: Our investigation encompasses tumor geometry and surrounding tissue, characterized by well-defined boundaries and initial conditions. Utilizing the finite element method, we solve governing equations across a range of parameters: dissociation constant KD (1, 0.1, 0.01 [nM]), internalization rate (0.01-0.0001 [min-1]), diverse tumor shapes, and variable necrotic zone sizes. This model can provide an accurate analysis of radiopharmaceutical delivery from the injection site to the tumor cell, including drug transport in the vascular, interstitial, and intracellular spaces, and considering important parameters (e.g., drug extravasation from microvessels or to lymphatic vessels, the extracellular matrix, receptors, and intracellular space). RESULTS: Our findings reveal significant enhancements in tumor-absorbed doses as KD decreases. This outcome can be attributed to the higher affinity of radiopharmaceuticals for PSMA receptors as KD diminishes, facilitating a more efficient binding and retention of the therapeutic agent within the tumor microenvironment. Additionally, tumor-absorbed doses for KD ∼ 1 [nM] show an upward trend with higher internalization rates. This observation can be rationalized by considering that a greater internalization rate would result in a higher proportion of radiopharmaceuticals being taken up by tumor cells after binding to receptors on the cell surface. Notably, tumor shape and necrotic zone size exhibit limited influence on tumor absorbed dose. CONCLUSIONS: The present study employs the CDR model to explore the role of physiological parameters in shaping 177Lu-PSMA-617 RPT delivery. These findings provide insights for improving prostate cancer therapy by understanding radiopharmaceutical transport dynamics. This computational approach contributes to advancing our understanding of radiopharmaceutical delivery mechanisms and has implications for enhancing treatment efficacy.


Prostatic Neoplasms , Radiopharmaceuticals , Male , Humans , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/chemistry , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/radiotherapy , Dipeptides/therapeutic use , Dipeptides/chemistry , Tumor Microenvironment
20.
Med Phys ; 51(2): 1203-1216, 2024 Feb.
Article En | MEDLINE | ID: mdl-37544015

BACKGROUND: Prostate-specific membrane antigen (PSMA) PET imaging represents a valuable source of information reflecting disease stage, response rate, and treatment optimization options, particularly with PSMA radioligand therapy. Quantification of radiopharmaceutical uptake in healthy organs from PSMA images has the potential to minimize toxicity by extrapolation of the radiation dose delivery towards personalization of therapy. However, segmentation and quantification of uptake in organs requires labor-intensive organ delineations that are often not feasible in the clinic nor scalable for large clinical trials. PURPOSE: In this work we develop and test the PSMA Healthy organ segmentation network (PSMA-Hornet), a fully-automated deep neural net for simultaneous segmentation of 14 healthy organs representing the normal biodistribution of [18 F]DCFPyL on PET/CT images. We also propose a modified U-net architecture, a self-supervised pre-training method for PET/CT images, a multi-target Dice loss, and multi-target batch balancing to effectively train PSMA-Hornet and similar networks. METHODS: The study used manually-segmented [18 F]DCFPyL PET/CT images from 100 subjects, and 526 similar images without segmentations. The unsegmented images were used for self-supervised model pretraining. For supervised training, Monte-Carlo cross-validation was used to evaluate the network performance, with 85 subjects in each trial reserved for model training, 5 for validation, and 10 for testing. Image segmentation and quantification metrics were evaluated on the test folds with respect to manual segmentations by a nuclear medicine physician, and compared to inter-rater agreement. The model's segmentation performance was also evaluated on a separate set of 19 images with high tumor load. RESULTS: With our best model, the lowest mean Dice coefficient on the test set was 0.826 for the sublingual gland, and the highest was 0.964 for liver. The highest mean error in tracer uptake quantification was 13.9% in the sublingual gland. Self-supervised pretraining improved training convergence, train-to-test generalization, and segmentation quality. In addition, we found that a multi-target network produced significantly higher segmentation accuracy than single-organ networks. CONCLUSIONS: The developed network can be used to automatically obtain high-quality organ segmentations for PSMA image analysis tasks. It can be used to reproducibly extract imaging data, and holds promise for clinical applications such as personalized radiation dosimetry and improved radioligand therapy.


Antigens, Surface , Glutamate Carboxypeptidase II , Prostatic Neoplasms , Animals , Humans , Male , Image Processing, Computer-Assisted/methods , Positron Emission Tomography Computed Tomography/methods , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/radiotherapy , Tissue Distribution
...