Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
1.
Radiol Oncol ; 58(3): 335-347, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39287171

RESUMO

BACKGROUND: To evaluate the role of the novel quantitative imaging biomarker (QIB) SUVX% of 18F-FDG uptake extracted from early 18F-FDG-PET/CT scan at 4 weeks for the detection of immune-related adverse events (rAE) in a cohort of patients with metastatic melanoma (mM) patients receiving immune-checkpoint inhibitors (ICI). PATIENTS AND METHODS: In this prospective non-interventional, one-centre clinical study, patients with mM, receiving ICI treatment, were regularly followed by 18F-FDG PET/CT. Patients were scanned at baseline, early point at week four (W4), week sixteen (W16) and week thirty-two (W32) after ICI initiation. A convolutional neural network (CNN) was used to segment three organs: lung, bowel, thyroid. QIB of irAE - SUVX% - was analyzed within the target organs and correlated with the clinical irAE status. Area under the receiver-operating characteristic curve (AUROC) was used to quantify irAE detection performance. RESULTS: A total of 242 18F-FDG PET/CT images of 71 mM patients were prospectively collected and analysed. The early W4 scan showed improved detection only for the thyroid gland compared to W32 scan (p=0.047). The AUROC for detection of irAE in the three target organs was highest when SUVX% was extracted from W16 scan and was 0.76 for lung, 0.53 for bowel and 0.81 for thyroid. SUVX% extracted from W4 scan did not improve detection of irAE compared to W16 scan (lung: p = 0.54, bowel: p = 0.75, thyroid: p = 0.3, DeLong test), as well as compared to W32 scan in lungs (p = 0.32) and bowel (p = 0.3). CONCLUSIONS: Early time point 18F-FDG PET/CT at W4 did not lead to statistically significant earlier detection of irAE. However, organ 18F-FDG uptake as quantified by SUVX% proved to be a consistent QIB of irAE. To better assess the role of 18F-FDG PET/CT in irAE detection, the time evolution of 18F-FDG PET/CT quantifiable inflammation would be of essence, only achievable in multi centric studies.


Assuntos
Fluordesoxiglucose F18 , Inibidores de Checkpoint Imunológico , Melanoma , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Compostos Radiofarmacêuticos , Humanos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Melanoma/diagnóstico por imagem , Melanoma/imunologia , Estudos Prospectivos , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Inibidores de Checkpoint Imunológico/efeitos adversos , Adulto , Neoplasias Cutâneas/diagnóstico por imagem , Neoplasias Cutâneas/patologia , Curva ROC , Glândula Tireoide/diagnóstico por imagem
2.
Phys Med Biol ; 69(15)2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-38981594

RESUMO

Objective.Deep learning models that aid in medical image assessment tasks must be both accurate and reliable to be deployed within clinical settings. While deep learning models have been shown to be highly accurate across a variety of tasks, measures that indicate the reliability of these models are less established. Increasingly, uncertainty quantification (UQ) methods are being introduced to inform users on the reliability of model outputs. However, most existing methods cannot be augmented to previously validated models because they are not post hoc, and they change a model's output. In this work, we overcome these limitations by introducing a novel post hoc UQ method, termedLocal Gradients UQ, and demonstrate its utility for deep learning-based metastatic disease delineation.Approach.This method leverages a trained model's localized gradient space to assess sensitivities to trained model parameters. We compared the Local Gradients UQ method to non-gradient measures defined using model probability outputs. The performance of each uncertainty measure was assessed in four clinically relevant experiments: (1) response to artificially degraded image quality, (2) comparison between matched high- and low-quality clinical images, (3) false positive (FP) filtering, and (4) correspondence with physician-rated disease likelihood.Main results.(1) Response to artificially degraded image quality was enhanced by the Local Gradients UQ method, where the median percent difference between matching lesions in non-degraded and most degraded images was consistently higher for the Local Gradients uncertainty measure than the non-gradient uncertainty measures (e.g. 62.35% vs. 2.16% for additive Gaussian noise). (2) The Local Gradients UQ measure responded better to high- and low-quality clinical images (p< 0.05 vsp> 0.1 for both non-gradient uncertainty measures). (3) FP filtering performance was enhanced by the Local Gradients UQ method when compared to the non-gradient methods, increasing the area under the receiver operating characteristic curve (ROC AUC) by 20.1% and decreasing the false positive rate by 26%. (4) The Local Gradients UQ method also showed more favorable correspondence with physician-rated likelihood for malignant lesions by increasing ROC AUC for correspondence with physician-rated disease likelihood by 16.2%.Significance. In summary, this work introduces and validates a novel gradient-based UQ method for deep learning-based medical image assessments to enhance user trust when using deployed clinical models.


Assuntos
Aprendizado Profundo , Processamento de Imagem Assistida por Computador , Incerteza , Humanos , Processamento de Imagem Assistida por Computador/métodos
3.
Eur J Nucl Med Mol Imaging ; 51(11): 3428-3439, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38795121

RESUMO

PURPOSE: Somatostatin receptor (SSTR) imaging features are predictive of treatment outcome for neuroendocrine tumor (NET) patients receiving peptide receptor radionuclide therapy (PRRT). However, comprehensive (all metastatic lesions), longitudinal (temporal variation), and lesion-level measured features have never been explored. Such features allow for capturing the heterogeneity in disease response to treatment. Furthermore, models combining these features are lacking. In this work we evaluated the predictive power of comprehensive, longitudinal, lesion-level 68GA-SSTR-PET features combined with a multivariate linear regression (MLR) model. METHODS: This retrospective study enrolled NET patients treated with [177Lu]Lu-DOTA-TATE and imaged with [68Ga]Ga-DOTA-TATE at baseline and post-therapy. All lesions were segmented, anatomically labeled, and longitudinally matched. Lesion-level uptake and variation in uptake were measured. Patient-level features were engineered and selected for modeling of progression-free survival (PFS). The model was validated via concordance index, patient classification (ROC analysis), and survival analysis (Kaplan-Meier and Cox proportional hazards). The MLR was benchmarked against single feature predictions. RESULTS: Thirty-six NET patients were enrolled and stratified into poor and good responders (PFS ≥ 25 months). Four patient-level features were selected, the MLR concordance index was 0.826, and the AUC was 0.88 (0.85 specificity, 0.81 sensitivity). Survival analysis led to significant patient stratification (p<.001) and hazard ratio (3⨯10-5). Lastly, in a benchmark study, the MLR modeling approach outperformed all the single feature predictors. CONCLUSION: Comprehensive, lesion-level, longitudinal 68GA-SSTR-PET analysis, combined with MLR modeling, leads to excellent predictions of PRRT outcome in NET patients, outperforming non-comprehensive, patient-level, and single time-point feature predictions. MESSAGE: Neuroendocrine tumor, peptide receptor radionuclide therapy, Somatostatin Receptor Imaging, Outcome Prediction, Treatment Response Assessment.


Assuntos
Tumores Neuroendócrinos , Octreotida , Compostos Organometálicos , Humanos , Tumores Neuroendócrinos/diagnóstico por imagem , Tumores Neuroendócrinos/radioterapia , Masculino , Feminino , Pessoa de Meia-Idade , Octreotida/análogos & derivados , Octreotida/uso terapêutico , Compostos Organometálicos/uso terapêutico , Idoso , Estudos Retrospectivos , Resultado do Tratamento , Adulto , Tomografia por Emissão de Pósitrons , Prognóstico , Estudos Longitudinais
4.
Artigo em Inglês | MEDLINE | ID: mdl-38819668

RESUMO

PURPOSE: Standardized reporting of treatment response in oncology patients has traditionally relied on methods like RECIST, PERCIST and Deauville score. These endpoints assess only a few lesions, potentially overlooking the response heterogeneity of all disease. This study hypothesizes that comprehensive spatial-temporal evaluation of all individual lesions is necessary for superior prognostication of clinical outcome. METHODS: [18F]FDG PET/CT scans from 241 patients (127 diffuse large B-cell lymphoma (DLBCL) and 114 non-small cell lung cancer (NSCLC)) were retrospectively obtained at baseline and either during chemotherapy or post-chemoradiotherapy. An automated TRAQinform IQ software (AIQ Solutions) analyzed the images, performing quantification of change in regions of interest suspicious of cancer (lesion-ROI). Multivariable Cox proportional hazards (CoxPH) models were trained to predict overall survival (OS) with varied sets of quantitative features and lesion-ROI, compared by bootstrapping with C-index and t-tests. The best-fit model was compared to automated versions of previously established methods like RECIST, PERCIST and Deauville score. RESULTS: Multivariable CoxPH models demonstrated superior prognostic power when trained with features quantifying response heterogeneity in all individual lesion-ROI in DLBCL (C-index = 0.84, p < 0.001) and NSCLC (C-index = 0.71, p < 0.001). Prognostic power significantly deteriorated (p < 0.001) when using subsets of lesion-ROI (C-index = 0.78 and 0.67 for DLBCL and NSCLC, respectively) or excluding response heterogeneity (C-index = 0.67 and 0.70). RECIST, PERCIST, and Deauville score could not significantly associate with OS (C-index < 0.65 and p > 0.1), performing significantly worse than the multivariable models (p < 0.001). CONCLUSIONS: Quantitative evaluation of response heterogeneity of all individual lesions is necessary for the superior prognostication of clinical outcome.

5.
Phys Med Biol ; 69(11)2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38749468

RESUMO

Objective.Fast and accurate deformable image registration (DIR), including DIR uncertainty estimation, is essential for safe and reliable clinical deployment. While recent deep learning models have shown promise in predicting DIR with its uncertainty, challenges persist in proper uncertainty evaluation and hyperparameter optimization for these methods. This work aims to develop and evaluate a model that can perform fast DIR and predict its uncertainty in seconds.Approach.This study introduces a novel probabilistic multi-resolution image registration model utilizing convolutional neural networks to estimate a multivariate normal distributed dense displacement field (DDF) in a multimodal image registration problem. To assess the quality of the DDF distribution predicted by the model, we propose a new metric based on the Kullback-Leibler divergence. The performance of our approach was evaluated against three other DIR algorithms (VoxelMorph, Monte Carlo dropout, and Monte Carlo B-spline) capable of predicting uncertainty. The evaluation of the models included not only the quality of the deformation but also the reliability of the estimated uncertainty. Our application investigated the registration of a treatment planning computed tomography (CT) to follow-up cone beam CT for daily adaptive radiotherapy.Main results.The hyperparameter tuning of the models showed a trade-off between the estimated uncertainty's reliability and the deformation's accuracy. In the optimal trade-off, our model excelled in contour propagation and uncertainty estimation (p <0.05) compared to existing uncertainty estimation models. We obtained an average dice similarity coefficient of 0.89 and a KL-divergence of 0.15.Significance.By addressing challenges in DIR uncertainty estimation and evaluation, our work showed that both the DIR and its uncertainty can be reliably predicted, paving the way for safe deployment in a clinical environment.


Assuntos
Processamento de Imagem Assistida por Computador , Redes Neurais de Computação , Incerteza , Processamento de Imagem Assistida por Computador/métodos , Humanos , Algoritmos , Planejamento da Radioterapia Assistida por Computador/métodos , Tomografia Computadorizada de Feixe Cônico/métodos
6.
Phys Med Biol ; 69(8)2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38457838

RESUMO

Objective. Manual analysis of individual cancer lesions to assess disease response is clinically impractical and requires automated lesion tracking methodologies. However, no methodology has been developed for whole-body individual lesion tracking, across an arbitrary number of scans, and acquired with various imaging modalities.Approach. This study introduces a lesion tracking methodology and benchmarked it using 2368Ga-DOTATATE PET/CT and PET/MR images of eight neuroendocrine tumor patients. The methodology consists of six steps: (1) alignment of multiple scans via image registration, (2) body-part labeling, (3) automatic lesion-wise dilation, (4) clustering of lesions based on local lesion shape metrics, (5) assignment of lesion tracks, and (6) output of a lesion graph. Registration performance was evaluated via landmark distance, lesion matching accuracy was evaluated between each image pair, and lesion tracking accuracy was evaluated via identical track ratio. Sensitivity studies were performed to evaluate the impact of lesion dilation (fixed versus automatic dilation), anatomic location, image modalities (inter- versus intra-modality), registration mode (direct versus indirect registration), and track size (number of time-points and lesions) on lesion matching and tracking performance.Main results. Manual contouring yielded 956 lesions, 1570 lesion-matching decisions, and 493 lesion tracks. The median residual registration error was 2.5 mm. The automatic lesion dilation led to 0.90 overall lesion matching accuracy, and an 88% identical track ratio. The methodology is robust regarding anatomic locations, image modalities, and registration modes. The number of scans had a moderate negative impact on the identical track ratio (94% for 2 scans, 91% for 3 scans, and 81% for 4 scans). The number of lesions substantially impacted the identical track ratio (93% for 2 nodes versus 54% for ≥5 nodes).Significance. The developed methodology resulted in high lesion-matching accuracy and enables automated lesion tracking in PET/CT and PET/MR.


Assuntos
Tumores Neuroendócrinos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Humanos , Tomografia Computadorizada por Raios X/métodos , Imagem Multimodal/métodos , Tomografia por Emissão de Pósitrons/métodos , Tumores Neuroendócrinos/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos
7.
Br J Radiol ; 96(1152): 20221178, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37751168

RESUMO

OBJECTIVE: This study aimed to quantify both the intra- and intertracer repeatability of lesion-level radiomics features in [68Ga]Ga-prostate-specific membrane antigen (PSMA)-11 and [18F]F-PSMA-1007 positron emission tomography (PET) scans. METHODS: Eighteen patients with metastatic prostate cancer (mPCa) were prospectively recruited for the study and randomised to one of three test-retest groups: (i) intratracer [68Ga]Ga-PSMA-11 PET, (ii) intratracer [18F]F-PSMA-1007 PET or (iii) intertracer between [68Ga]Ga-PSMA-11 and [18F]F-PSMA-1007 PET. Four conventional PET metrics (standardised uptake value (SUV)max, SUVmean, SUVtotal and volume) and 107 radiomics features were extracted from 75 lesions and assessed using the repeatability coefficient (RC) and the ICC. Radiomic feature repeatability was also quantified after the application of 16 filters to the PET image. RESULTS: Test-retest scans were taken a median of 5 days apart (range: 2-7 days). SUVmean demonstrated the lowest RC limits of the conventional features, with RCs of 7.9%, 14.2% and 24.7% for the [68Ga]Ga-PSMA-11 PET, [18F]F-PSMA-1007 PET, and intertracer groups, respectively. 69%, 66% and 9% of all radiomics features had good or excellent ICC values (ICC ≥ 0.75) for the same groups. Feature repeatability therefore diminished considerably for the intertracer group relative to intratracer groups. CONCLUSION: In this study, robust biomarkers for each tracer group that can be used in subsequent clinical studies were identified. Overall, the repeatability of conventional and radiomic features were found to be substantially lower for the intertracer group relative to both intratracer groups, suggesting that assessing patient response quantitatively should be done using the same radiotracer where possible. ADVANCES IN KNOWLEDGE: Intertracer biomarker repeatability limits are significantly larger than intratracer limits.


Assuntos
Radioisótopos de Gálio , Neoplasias da Próstata , Masculino , Humanos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Estudos Prospectivos , Radiômica , Tomografia por Emissão de Pósitrons , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/patologia
8.
Phys Med Biol ; 68(17)2023 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-37567220

RESUMO

Objective.Patients with metastatic disease are followed throughout treatment with medical imaging, and accurately assessing changes of individual lesions is critical to properly inform clinical decisions. The goal of this work was to assess the performance of an automated lesion-matching algorithm in comparison to inter-reader variability (IRV) of matching lesions between scans of metastatic cancer patients.Approach.Forty pairs of longitudinal PET/CT and CT scans were collected and organized into four cohorts: lung cancers, head and neck cancers, lymphomas, and advanced cancers. Cases were also divided by cancer burden: low-burden (<10 lesions), intermediate-burden (10-29), and high-burden (30+). Two nuclear medicine physicians conducted independent reviews of each scan-pair and manually matched lesions. Matching differences between readers were assessed to quantify the IRV of lesion matching. The two readers met to form a consensus, which was considered a gold standard and compared against the output of an automated lesion-matching algorithm. IRV and performance of the automated method were quantified using precision, recall, F1-score, and the number of differences.Main results.The performance of the automated method did not differ significantly from IRV for any metric in any cohort (p> 0.05, Wilcoxon paired test). In high-burden cases, the F1-score (median [range]) was 0.89 [0.63, 1.00] between the automated method and reader consensus and 0.93 [0.72, 1.00] between readers. In low-burden cases, F1-scores were 1.00 [0.40, 1.00] and 1.00 [0.40, 1.00], for the automated method and IRV, respectively. Automated matching was significantly more efficient than either reader (p< 0.001). In high-burden cases, median matching time for the readers was 60 and 30 min, respectively, while automated matching took a median of 3.9 minSignificance.The automated lesion-matching algorithm was successful in performing lesion matching, meeting the benchmark of IRV. Automated lesion matching can significantly expedite and improve the consistency of longitudinal lesion-matching.


Assuntos
Neoplasias Pulmonares , Linfoma , Humanos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Tomografia Computadorizada por Raios X/métodos , Algoritmos
9.
Phys Med Biol ; 68(11)2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-37137317

RESUMO

Objective. Deep Learning models are often susceptible to failures after deployment. Knowing when your model is producing inadequate predictions is crucial. In this work, we investigate the utility of Monte Carlo (MC) dropout and the efficacy of the proposed uncertainty metric (UM) for flagging of unacceptable pectoral muscle segmentations in mammograms.Approach. Segmentation of pectoral muscle was performed with modified ResNet18 convolutional neural network. MC dropout layers were kept unlocked at inference time. For each mammogram, 50 pectoral muscle segmentations were generated. The mean was used to produce the final segmentation and the standard deviation was applied for the estimation of uncertainty. From each pectoral muscle uncertainty map, the overall UM was calculated. To validate the UM, a correlation between the dice similarity coefficient (DSC) and UM was used. The UM was first validated in a training set (200 mammograms) and finally tested in an independent dataset (300 mammograms). ROC-AUC analysis was performed to test the discriminatory power of the proposed UM for flagging unacceptable segmentations.Main results. The introduction of dropout layers in the model improved segmentation performance (DSC = 0.95 ± 0.07 versus DSC = 0.93 ± 0.10). Strong anti-correlation (r= -0.76,p< 0.001) between the proposed UM and DSC was observed. A high AUC of 0.98 (97% specificity at 100% sensitivity) was obtained for the discrimination of unacceptable segmentations. Qualitative inspection by the radiologist revealed that images with high UM are difficult to segment.Significance. The use of MC dropout at inference time in combination with the proposed UM enables flagging of unacceptable pectoral muscle segmentations from mammograms with excellent discriminatory power.


Assuntos
Aprendizado Profundo , Músculos Peitorais/diagnóstico por imagem , Incerteza , Redes Neurais de Computação , Mamografia/métodos , Processamento de Imagem Assistida por Computador/métodos
10.
Ecancermedicalscience ; 17: 1508, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37113724

RESUMO

The rising cancer incidence and mortality in sub-Saharan Africa (SSA) warrants an increased focus on adopting or developing approaches that can significantly increase access to treatment in the region. One such approach recommended by the recent Lancet Oncology Commission for sub-Saharan Africa is hypofractionated radiotherapy (HFRT), which can substantially increase access to radiotherapy by reducing the overall duration of time (in days) each person spends being treated. Here we highlight challenges in adopting such an approach identified during the implementation of the HypoAfrica clinical trial. The HypoAfrica clinical trial is a longitudinal, multicentre study exploring the feasibility of applying HFRT for prostate cancer in SSA. This study has presented an opportunity for a pragmatic assessment of potential barriers and facilitators to adopting HFRT. Our results highlight three key challenges: quality assurance, study harmonisation and machine maintenance. We describe solutions employed to resolve these challenges and opportunities for longer term solutions that can facilitate scaling-up use of HFRT in SSA in clinical care and multicentre clinical trials. This report provides a valuable reference for the utilisation of radiotherapy approaches that increase access to treatment and the conduct of high-quality large-scale/multi-centre clinical trials involving radiotherapy. Trial registration: Not available yet.

12.
13.
Phys Med Biol ; 68(3)2023 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-36580684

RESUMO

Objective.Manual disease delineation in full-body imaging of patients with multiple metastases is often impractical due to high disease burden. However, this is a clinically relevant task as quantitative image techniques assessing individual metastases, while limited, have been shown to be predictive of treatment outcome. The goal of this work was to evaluate the efficacy of deep learning-based methods for full-body delineation of skeletal metastases and to compare their performance to existing methods in terms of disease delineation accuracy and prognostic power.Approach.1833 suspicious lesions on 3718F-NaF PET/CT scans of patients with metastatic castration-resistant prostate cancer (mCRPC) were contoured and classified as malignant, equivocal, or benign by a nuclear medicine physician. Two convolutional neural network (CNN) architectures (DeepMedic and nnUNet)were trained to delineate malignant disease regions with and without three-model ensembling. Malignant disease contours using previously established methods were obtained. The performance of each method was assessed in terms of four different tasks: (1) detection, (2) segmentation, (3) PET SUV metric correlations with physician-based data, and (4) prognostic power of progression-free survival.Main Results.The nnUnet three-model ensemble achieved superior detection performance with a mean (+/- standard deviation) sensitivity of 82.9±ccc 0.1% at the selected operating point. The nnUnet single and three-model ensemble achieved comparable segmentation performance with a mean Dice coefficient of 0.80±0.12 and 0.79±0.12, respectively, both outperforming other methods. The nnUNet ensemble achieved comparable or superior SUV metric correlation performance to gold-standard data. Despite superior disease delineation performance, the nnUNet methods did not display superior prognostic power over other methods.Significance.This work showed that CNN-based (nnUNet) methods are superior to the non-CNN methods for mCRPC disease delineation in full-body18F-NaF PET/CT. The CNN-based methods, however, do not hold greater prognostic power for predicting clinical outcome. This merits more investigation on the optimal selection of delineation methods for specific clinical tasks.


Assuntos
Neoplasias Ósseas , Neoplasias de Próstata Resistentes à Castração , Masculino , Humanos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Neoplasias de Próstata Resistentes à Castração/patologia , Prognóstico , Neoplasias Ósseas/diagnóstico por imagem , Neoplasias Ósseas/secundário , Cintilografia
14.
Nat Commun ; 13(1): 7346, 2022 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-36470898

RESUMO

Although machine learning (ML) has shown promise across disciplines, out-of-sample generalizability is concerning. This is currently addressed by sharing multi-site data, but such centralization is challenging/infeasible to scale due to various limitations. Federated ML (FL) provides an alternative paradigm for accurate and generalizable ML, by only sharing numerical model updates. Here we present the largest FL study to-date, involving data from 71 sites across 6 continents, to generate an automatic tumor boundary detector for the rare disease of glioblastoma, reporting the largest such dataset in the literature (n = 6, 314). We demonstrate a 33% delineation improvement for the surgically targetable tumor, and 23% for the complete tumor extent, over a publicly trained model. We anticipate our study to: 1) enable more healthcare studies informed by large diverse data, ensuring meaningful results for rare diseases and underrepresented populations, 2) facilitate further analyses for glioblastoma by releasing our consensus model, and 3) demonstrate the FL effectiveness at such scale and task-complexity as a paradigm shift for multi-site collaborations, alleviating the need for data-sharing.


Assuntos
Big Data , Glioblastoma , Humanos , Aprendizado de Máquina , Doenças Raras , Disseminação de Informação
16.
Phys Med Biol ; 67(19)2022 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-36055243

RESUMO

Objective. Neuroimaging uncovers important information about disease in the brain. Yet in Alzheimer's disease (AD), there remains a clear clinical need for reliable tools to extract diagnoses from neuroimages. Significant work has been done to develop deep learning (DL) networks using neuroimaging for AD diagnosis. However, no particular model has emerged as optimal. Due to a lack of direct comparisons and evaluations on independent data, there is no consensus on which modality is best for diagnostic models or whether longitudinal information enhances performance. The purpose of this work was (1) to develop a generalizable DL model to distinguish neuroimaging scans of AD patients from controls and (2) to evaluate the influence of imaging modality and longitudinal data on performance.Approach. We trained a 2-class convolutional neural network (CNN) with and without a cascaded recurrent neural network (RNN). We used datasets of 772 (NAD = 364,Ncontrol= 408) 3D18F-FDG PET scans and 780 (NAD = 280,Ncontrol= 500) T1-weighted volumetric-3D MR images (containing 131 and 144 patients with multiple timepoints) from the Alzheimer's Disease Neuroimaging Initiative, plus an independent set of 104 (NAD = 63,NNC = 41)18F-FDG PET scans (one per patient) for validation.Main Results. ROC analysis showed that PET-trained models outperformed MRI-trained, achieving maximum AUC with the CNN + RNN model of 0.93 ± 0.08, with accuracy 82.5 ± 8.9%. Adding longitudinal information offered significant improvement to performance on18F-FDG PET, but not on T1-MRI. CNN model validation with an independent18F-FDG PET dataset achieved AUC of 0.99. Layer-wise relevance propagation heatmaps added CNN interpretability.Significance. The development of a high-performing tool for AD diagnosis, with the direct evaluation of key influences, reveals the advantage of using18F-FDG PET and longitudinal data over MRI and single timepoint analysis. This has significant implications for the potential of neuroimaging for future research on AD diagnosis and clinical management of suspected AD patients.


Assuntos
Doença de Alzheimer , Aprendizado Profundo , Doença de Alzheimer/diagnóstico por imagem , Fluordesoxiglucose F18 , Humanos , Imageamento por Ressonância Magnética/métodos , Neuroimagem/métodos , Tomografia por Emissão de Pósitrons/métodos
17.
Eur J Nucl Med Mol Imaging ; 49(6): 1857-1869, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34958422

RESUMO

PURPOSE: To develop quantitative molecular imaging biomarkers of immune-related adverse event (irAE) development in malignant melanoma (MM) patients receiving immune-checkpoint inhibitors (ICI) imaged with 18F-FDG PET/CT. METHODS: 18F-FDG PET/CT images of 58 MM patients treated with anti-PD-1 or anti-CTLA-4 ICI were retrospectively analyzed for indication of irAE. Three target organs, most commonly affected by irAE, were considered: bowel, lung, and thyroid. Patient charts were reviewed to identify which patients experienced irAE, irAE grade, and time to irAE diagnosis. Target organs were segmented using a convolutional neural network (CNN), and novel quantitative imaging biomarkers - SUV percentiles (SUVX%) of 18F-FDG uptake within the target organs - were correlated with the clinical irAE status. Area under the receiver-operating characteristic curve (AUROC) was used to quantify irAE detection performance. Patients who did not experience irAE were used to establish normal ranges for target organ 18F-FDG uptake. RESULTS: A total of 31% (18/58) patients experienced irAE in the three target organs: bowel (n=6), lung (n=5), and thyroid (n=9). Optimal percentiles for identifying irAE were bowel (SUV95%, AUROC=0.79), lung (SUV95%, AUROC=0.98), and thyroid (SUV75%, AUROC=0.88). Optimal cut-offs for irAE detection were bowel (SUV95%>2.7 g/mL), lung (SUV95%>1.7 g/mL), and thyroid (SUV75%>2.1 g/mL). Normal ranges (95% confidence interval) for the SUV percentiles in patients without irAE were bowel [1.74, 2.86 g/mL], lung [0.73, 1.46 g/mL], and thyroid [0.86, 1.99 g/mL]. CONCLUSIONS: Increased 18F-FDG uptake within irAE-affected organs provides predictive information about the development of irAE in MM patients receiving ICI and represents a potential quantitative imaging biomarker for irAE. Some irAE can be detected on 18F-FDG PET/CT well before clinical symptoms appear.


Assuntos
Melanoma , Segunda Neoplasia Primária , Biomarcadores , Fluordesoxiglucose F18 , Humanos , Inibidores de Checkpoint Imunológico/efeitos adversos , Melanoma/diagnóstico por imagem , Melanoma/tratamento farmacológico , Projetos Piloto , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Tomografia por Emissão de Pósitrons , Estudos Retrospectivos , Neoplasias Cutâneas , Melanoma Maligno Cutâneo
18.
Front Oncol ; 11: 771787, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34790581

RESUMO

Metastatic Prostate Cancer (mPCa) is associated with a poor patient prognosis. mPCa spreads throughout the body, often to bones, with spatial and temporal variations that make the clinical management of the disease difficult. The evolution of the disease leads to spatial heterogeneity that is extremely difficult to characterise with solid biopsies. Imaging provides the opportunity to quantify disease spread. Advanced image analytics methods, including radiomics, offer the opportunity to characterise heterogeneity beyond what can be achieved with simple assessment. Radiomics analysis has the potential to yield useful quantitative imaging biomarkers that can improve the early detection of mPCa, predict disease progression, assess response, and potentially inform the choice of treatment procedures. Traditional radiomics analysis involves modelling with hand-crafted features designed using significant domain knowledge. On the other hand, artificial intelligence techniques such as deep learning can facilitate end-to-end automated feature extraction and model generation with minimal human intervention. Radiomics models have the potential to become vital pieces in the oncology workflow, however, the current limitations of the field, such as limited reproducibility, are impeding their translation into clinical practice. This review provides an overview of the radiomics methodology, detailing critical aspects affecting the reproducibility of features, and providing examples of how artificial intelligence techniques can be incorporated into the workflow. The current landscape of publications utilising radiomics methods in the assessment and treatment of mPCa are surveyed and reviewed. Associated studies have incorporated information from multiple imaging modalities, including bone scintigraphy, CT, PET with varying tracers, multiparametric MRI together with clinical covariates, spanning the prediction of progression through to overall survival in varying cohorts. The methodological quality of each study is quantified using the radiomics quality score. Multiple deficits were identified, with the lack of prospective design and external validation highlighted as major impediments to clinical translation. These results inform some recommendations for future directions of the field.

19.
Phys Med Biol ; 66(21)2021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-34644696

RESUMO

Intro.Current radiation therapy (RT) planning guidelines handle uncertainties in RT using geometric margins. This approach is simple to use but oversimplifies complex underlying processes and is cumbersome for non-homogeneous dose prescriptions. In this work, we characterize the performance of a novel probabilistic target definition and planning (PTP) approach, which uses voxel-level tumor likelihood information in treatment plan optimization.Methods.We expanded a treatment planning system with probabilistic therapy planning functionality that utilizes non-binary target maps (TM) as voxel-level input to dose plan optimization. Different dose plans were calculated and compared for twelve prostate cancer patients with multiparametric magnetic resonance imaging derived TMs. Dose plans were created using both classical and PTP approaches for uniform and integrated dose boost prescriptions. Dose performance between the different approaches was compared using dose benchmarks on target and organ-at-risk (OAR) volumes.Results.Over all dose metrics, PTP was shown to be comparable to classical planning. For plans of uniform dose prescription, the PTP approach created plans within 1 Gy of the classical planning approach across all dose metrics, with no significant differences (p > 0.2). For plans with the integrated dose boost, PTP plans exhibited higher dose heterogeneity, but still showed target doses comparable to the classical approach, without increasing doses to OAR.Conclusion.In this work we introduce direct incorporation of probabilistic target definition into treatment planning. This treatment planning approach can produce both uniform dose plans and plans with integrated dose boosts that are comparable to ones created using classical dose planning. PTP is a flexible way to optimize external beam radiotherapy, as it is not limited by the use of margins. PTP can produce dose plans equivalent to classical planning, while also allows for greater versatility in dose prescription and direct incorporation of patient target definition uncertainty into treatment planning.


Assuntos
Imageamento por Ressonância Magnética Multiparamétrica , Neoplasias da Próstata , Radioterapia de Intensidade Modulada , Humanos , Masculino , Órgãos em Risco , Probabilidade , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/radioterapia , Dosagem Radioterapêutica , Planejamento da Radioterapia Assistida por Computador/métodos , Radioterapia de Intensidade Modulada/métodos
20.
Biomed Phys Eng Express ; 7(6)2021 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-34534974

RESUMO

Purpose.To investigate image intensity histograms as a potential source of useful imaging biomarkers in both a clinical example of detecting immune-related colitis (irColitis) in18F-FDG PET/CT images of immunotherapy patients and an idealized case of classifying digital reference objects (DRO).Methods.Retrospective analysis of bowel18F-FDG uptake in N = 40 patients receiving immune checkpoint inhibitors was conducted. A CNN trained to segment the bowel was used to generate the histogram of bowel18F-FDG uptake, and percentiles of the histogram were considered as potential metrics for detecting inflammation associated with irColitis. A model of the colon was also considered using cylindrical DRO. Classification of DRO with different intensity distributions was undertaken under varying geometry and noise settings.Results.The most predictive biomarker of irColitis was the 95th percentile of the bowel SUV histogram (SUV95%). Patients later diagnosed with irColitis had a significantly higher increase in SUV95%from baseline to first on-treatment PET than patients who did not experience irColitis (p = 0.02). An increase in SUV95%> + 40% separated pre-irColitis change from normal variability with a sensitivity of 75% and specificity of 88%. Furthermore, histogram percentiles were ideal metrics for classifying 'hot center' and 'cold center' DRO, and were robust to varying DRO geometry and noise, and to the presence of spoiler volumes unrelated to the detection task.Conclusions.The 95th percentile of the bowel SUV histogram was the optimal metric for detecting irColitis on18F-FDG PET/CT. Image intensity histograms are a promising source of imaging biomarkers for clinical tasks.


Assuntos
Colite , Fluordesoxiglucose F18 , Biomarcadores , Colite/diagnóstico , Humanos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA