Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Diagnostics (Basel) ; 14(9)2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38732298

RESUMO

Patlak slope (PS) images have the potential to improve lesion conspicuity compared with standardized uptake value (SUV) images but may be more artifact-prone. This study compared PS versus SUV image quality and hepatic tumor-to-background ratios (TBRs) at matched time points. Early and late SUV and PS images were reconstructed from dynamic positron emission tomography (PET) data. Two independent, blinded readers scored image quality metrics (a four-point Likert scale) and counted tracer-avid lesions. Hepatic lesions and parenchyma were segmented and quantitatively analyzed. Differences were assessed via the Wilcoxon signed-rank test (alpha, 0.05). Forty-three subjects were included. For overall quality and lesion detection, early PS images were significantly inferior to other reconstructions. For overall quality, late PS images (reader 1 [R1]: 3.95, reader 2 [R2]: 3.95) were similar (p > 0.05) to early SUV images (R1: 3.88, R2: 3.84) but slightly superior (p ≤ 0.002) to late SUV images (R1: 2.97, R2: 3.44). For lesion detection, late PS images were slightly inferior to late SUV images (R1 only) but slightly superior to early SUV images (both readers). PS-based TBRs were significantly higher than SUV-based TBRs at the early time point, with opposite findings at the late time point. In conclusion, late PS images are similar to early/late SUV images in image quality and lesion detection; the superiority of SUV versus PS hepatic TBRs is time-dependent.

2.
Mol Imaging Biol ; 26(2): 284-293, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38466523

RESUMO

PURPOSE: We aimed to determine the test-retest repeatability of quantitative metrics based on the Patlak slope (PS) versus the standardized uptake value (SUV) among lesions and normal organs on oncologic [18F]FDG-PET/CT. PROCEDURES: This prospective, single-center study enrolled adults undergoing standard-of-care oncologic [18F]FDG-PET/CTs. Early (35-50 min post-injection) and late (75-90 min post-injection) SUV and PS images were reconstructed from dynamic whole-body PET data. Repeat imaging occurred within 7 days. Relevant quantitative metrics were extracted from lesions and normal organs. Repeatability was assessed via mean test-retest percent changes [T-RT %Δ], within-subject coefficients of variation (wCVs), and intra-class correlation coefficients (ICCs). RESULTS: Nine subjects (mean age, 61.7 ± 6.2 years; 6 females) completed the test-retest protocol. Four subjects collectively had 17 [18F]FDG-avid lesions. Lesion wCVs were higher (i.e., worse repeatability) for PS-early-max (16.2%) and PS-early-peak (15.6%) than for SUV-early-max (8.9%) and SUV-early-peak (8.1%), with similar early metric ICCs (0.95-0.98). Lesion wCVs were similar for PS-late-max (8.5%) and PS-late-peak (6.4%) relative to SUV-late-max (9.7%) and SUV-late-peak (7.2%), with similar late metric ICCs (0.93-0.98). There was a significant bias toward higher retest SUV and PS values in the lesion analysis (T-RT %Δ [95% CI]: SUV-late-max, 10.0% [2.6%, 17.0%]; PS-late-max, 20.4% [14.3%, 26.4%]) but not in the normal organ analysis. CONCLUSIONS: Among [18F]FDG-avid lesions, the repeatability of PS-based metrics is similar to equivalent SUV-based metrics at late post-injection time points, indicating that PS-based metrics may be suitable for tracking response to oncologic therapies. However, further validation is required in light of our study's limitations, including small sample size and bias toward higher retest values for some metrics.


Assuntos
Fluordesoxiglucose F18 , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Adulto , Feminino , Humanos , Pessoa de Meia-Idade , Idoso , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Estudos Prospectivos , Reprodutibilidade dos Testes , Tomografia por Emissão de Pósitrons/métodos
3.
Cureus ; 15(8): e43343, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37700937

RESUMO

PURPOSE: Myocardial perfusion (MP) stress single-photon emission computed tomography (SPECT) is an established diagnostic test for patients suspected of coronary artery disease (CAD). Meanwhile, coronary artery calcification (CAC) scoring obtained from diagnostic CT is a highly sensitive test, offering incremental diagnostic information in identifying patients with significant CAD yet normal MP stress SPECT (MPSS) scans. However, after decades of wide utilization of MPSS, CAC is not commonly reimbursed (e.g. by the CMS), nor widely deployed in community settings. We studied the potential of complementary information deduced from the radiomics analysis of normal MPSS scans in predicting the CAC score. METHODS: We collected data from 428 patients with normal (non-ischemic) MPSS (99mTc-sestamibi; consensus reading). A nuclear medicine physician verified iteratively reconstructed images (attenuation-corrected) to be free from fixed perfusion defects and artifactual attenuation. Three-dimensional images were automatically segmented into four regions of interest (ROIs), including myocardium and three vascular segments (left anterior descending [LAD]-left circumference [LCX]-right coronary artery [RCA]). We used our software package, standardized environment for radiomics analysis (SERA), to extract 487 radiomic features in compliance with the image biomarker standardization initiative (IBSI). Isotropic cubic voxels were discretized using fixed bin-number discretization (eight schemes). We first performed blind-to-outcome feature selection focusing on a priori usefulness, dynamic range, and redundancy of features. Subsequently, we performed univariate and multivariate machine learning analyses to predict CAC scores from i) selected radiomic features, ii) 10 clinical features, and iii) combined radiomics + clinical features. Univariate analysis invoked Spearman correlation with Benjamini-Hotchberg false-discovery correction. The multivariate analysis incorporated stepwise linear regression, where we randomly selected a 15% test set and divided the other 85% of data into 70% training and 30% validation sets. Training started from a constant (intercept) model, iteratively adding/removing features (stepwise regression), invoking the Akaike information criterion (AIC) to discourage overfitting. Validation was run similarly, except that the training output model was used as the initial model. We randomized training/validation sets 20 times, selecting the best model using log-likelihood for evaluation in the test set. Assessment in the test set was performed thoroughly by running the entire operation 50 times, subsequently employing Fisher's method to verify the significance of independent tests. RESULTS: Unsupervised feature selection significantly reduced 8×487 features to 56. In univariate analysis, no feature survived the false-discovery rate (FDR) to directly correlate with CAC scores. Applying Fisher's method to the multivariate regression results demonstrated combining radiomics with the clinical features to enhance the significance of the prediction model across all cardiac segments.  Conclusions: Our standardized and statistically robust multivariate analysis demonstrated significant prediction of the CAC score for all cardiac segments when combining MPSS radiomic features with clinical features, suggesting radiomics analysis can add diagnostic or prognostic value to standard MPSS for wide clinical usage.

4.
EJNMMI Res ; 12(1): 76, 2022 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-36580220

RESUMO

BACKGROUND: Accurate classification of sites of interest on prostate-specific membrane antigen (PSMA) positron emission tomography (PET) images is an important diagnostic requirement for the differentiation of prostate cancer (PCa) from foci of physiologic uptake. We developed a deep learning and radiomics framework to perform lesion-level and patient-level classification on PSMA PET images of patients with PCa. METHODS: This was an IRB-approved, HIPAA-compliant, retrospective study. Lesions on [18F]DCFPyL PET/CT scans were assigned to PSMA reporting and data system (PSMA-RADS) categories and randomly partitioned into training, validation, and test sets. The framework extracted image features, radiomic features, and tissue type information from a cropped PET image slice containing a lesion and performed PSMA-RADS and PCa classification. Performance was evaluated by assessing the area under the receiver operating characteristic curve (AUROC). A t-distributed stochastic neighbor embedding (t-SNE) analysis was performed. Confidence and probability scores were measured. Statistical significance was determined using a two-tailed t test. RESULTS: PSMA PET scans from 267 men with PCa had 3794 lesions assigned to PSMA-RADS categories. The framework yielded AUROC values of 0.87 and 0.90 for lesion-level and patient-level PSMA-RADS classification, respectively, on the test set. The framework yielded AUROC values of 0.92 and 0.85 for lesion-level and patient-level PCa classification, respectively, on the test set. A t-SNE analysis revealed learned relationships between the PSMA-RADS categories and disease findings. Mean confidence scores reflected the expected accuracy and were significantly higher for correct predictions than for incorrect predictions (P < 0.05). Measured probability scores reflected the likelihood of PCa consistent with the PSMA-RADS framework. CONCLUSION: The framework provided lesion-level and patient-level PSMA-RADS and PCa classification on PSMA PET images. The framework was interpretable and provided confidence and probability scores that may assist physicians in making more informed clinical decisions.

5.
Cancers (Basel) ; 14(17)2022 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-36077686

RESUMO

Background: Prognostic risk factors for completely resected stage IA non-small-cell lung cancers (NSCLCs) have advanced minimally over recent decades. Although several biomarkers have been found to be associated with cancer recurrence, their added value to TNM staging and tumor grade are unclear. Methods: Features of preoperative low-dose CT image and histologic findings of hematoxylin- and eosin-stained tissue sections of resected lung tumor specimens were extracted from 182 stage IA NSCLC patients in the National Lung Screening Trial. These features were combined to predict the risk of tumor recurrence or progression through integrated deep learning evaluation (IDLE). Added values of IDLE to TNM staging and tumor grade in progression risk prediction and risk stratification were evaluated. Results: The 5-year AUC of IDLE was 0.817 ± 0.037 as compared to the AUC = 0.561 ± 0.042 and 0.573 ± 0.044 from the TNM stage and tumor grade, respectively. The IDLE score was significantly associated with cancer recurrence (p < 0.0001) even after adjusting for TNM staging and tumor grade. Synergy between chest CT image markers and histological markers was the driving force of the deep learning algorithm to produce a stronger prognostic predictor. Conclusions: Integrating markers from preoperative CT images and pathologist's readings of resected lung specimens through deep learning can improve risk stratification of stage 1A NSCLC patients over TNM staging and tumor grade alone. Our study suggests that combining markers from nonoverlapping platforms can increase the cancer risk prediction accuracy.

6.
Phys Med ; 89: 129-139, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34365117

RESUMO

PURPOSE: Positron emission tomography (PET) images tend to be significantly degraded by the partial volume effect (PVE) resulting from the limited spatial resolution of the reconstructed images. Our purpose is to propose a partial volume correction (PVC) method to tackle this issue. METHODS: In the present work, we explore a voxel-based PVC method under the least squares framework (LS) employing anatomical non-local means (NLMA) regularization. The well-known non-local means (NLM) filter utilizes the high degree of information redundancy that typically exists in images, and is typically used to directly reduce image noise by replacing each voxel intensity with a weighted average of its non-local neighbors. Here we explore NLM as a regularization term within iterative-deconvolution model to perform PVC. Further, an anatomical-guided version of NLM was proposed that incorporates MRI information into NLM to improve resolution and suppress image noise. The proposed approach makes subtle usage of the accompanying MRI information to define a more appropriate search space within the prior model. To optimize the regularized LS objective function, we used the Gauss-Seidel (GS) algorithm with the one-step-late (OSL) technique. RESULTS: After the import of NLMA, the visual and quality results are all improved. With a visual check, we notice that NLMA reduce the noise compared to other PVC methods. This is also validated in bias-noise curve compared to non-MRI-guided PVC framework. We can see NLMA gives better bias-noise trade-off compared to other PVC methods. CONCLUSIONS: Our efforts were evaluated in the base of amyloid brain PET imaging using the BrainWeb phantom and in vivo human data. We also compared our method with other PVC methods. Overall, we demonstrated the value of introducing subtle MRI-guidance in the regularization process, the proposed NLMA method resulting in promising visual as well as quantitative performance improvements.


Assuntos
Imageamento por Ressonância Magnética , Tomografia por Emissão de Pósitrons , Algoritmos , Encéfalo/diagnóstico por imagem , Humanos , Processamento de Imagem Assistida por Computador , Imagens de Fantasmas
7.
Phys Med Biol ; 65(24): 245032, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-32235059

RESUMO

An important need exists for reliable positron emission tomography (PET) tumor-segmentation methods for tasks such as PET-based radiation-therapy planning and reliable quantification of volumetric and radiomic features. To address this need, we propose an automated physics-guided deep-learning-based three-module framework to segment PET images on a per-slice basis. The framework is designed to help address the challenges of limited spatial resolution and lack of clinical training data with known ground-truth tumor boundaries in PET. The first module generates PET images containing highly realistic tumors with known ground-truth using a new stochastic and physics-based approach, addressing lack of training data. The second module trains a modified U-net using these images, helping it learn the tumor-segmentation task. The third module fine-tunes this network using a small-sized clinical dataset with radiologist-defined delineations as surrogate ground-truth, helping the framework learn features potentially missed in simulated tumors. The framework was evaluated in the context of segmenting primary tumors in 18F-fluorodeoxyglucose (FDG)-PET images of patients with lung cancer. The framework's accuracy, generalizability to different scanners, sensitivity to partial volume effects (PVEs) and efficacy in reducing the number of training images were quantitatively evaluated using Dice similarity coefficient (DSC) and several other metrics. The framework yielded reliable performance in both simulated (DSC: 0.87 (95% confidence interval (CI): 0.86, 0.88)) and patient images (DSC: 0.73 (95% CI: 0.71, 0.76)), outperformed several widely used semi-automated approaches, accurately segmented relatively small tumors (smallest segmented cross-section was 1.83 cm2), generalized across five PET scanners (DSC: 0.74 (95% CI: 0.71, 0.76)), was relatively unaffected by PVEs, and required low training data (training with data from even 30 patients yielded DSC of 0.70 (95% CI: 0.68, 0.71)). In conclusion, the proposed automated physics-guided deep-learning-based PET-segmentation framework yielded reliable performance in delineating tumors in FDG-PET images of patients with lung cancer.


Assuntos
Aprendizado Profundo , Processamento de Imagem Assistida por Computador/métodos , Neoplasias Pulmonares/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Automação , Humanos , Neoplasias Pulmonares/patologia
8.
Radiology ; 295(2): 328-338, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32154773

RESUMO

Background Radiomic features may quantify characteristics present in medical imaging. However, the lack of standardized definitions and validated reference values have hampered clinical use. Purpose To standardize a set of 174 radiomic features. Materials and Methods Radiomic features were assessed in three phases. In phase I, 487 features were derived from the basic set of 174 features. Twenty-five research teams with unique radiomics software implementations computed feature values directly from a digital phantom, without any additional image processing. In phase II, 15 teams computed values for 1347 derived features using a CT image of a patient with lung cancer and predefined image processing configurations. In both phases, consensus among the teams on the validity of tentative reference values was measured through the frequency of the modal value and classified as follows: less than three matches, weak; three to five matches, moderate; six to nine matches, strong; 10 or more matches, very strong. In the final phase (phase III), a public data set of multimodality images (CT, fluorine 18 fluorodeoxyglucose PET, and T1-weighted MRI) from 51 patients with soft-tissue sarcoma was used to prospectively assess reproducibility of standardized features. Results Consensus on reference values was initially weak for 232 of 302 features (76.8%) at phase I and 703 of 1075 features (65.4%) at phase II. At the final iteration, weak consensus remained for only two of 487 features (0.4%) at phase I and 19 of 1347 features (1.4%) at phase II. Strong or better consensus was achieved for 463 of 487 features (95.1%) at phase I and 1220 of 1347 features (90.6%) at phase II. Overall, 169 of 174 features were standardized in the first two phases. In the final validation phase (phase III), most of the 169 standardized features could be excellently reproduced (166 with CT; 164 with PET; and 164 with MRI). Conclusion A set of 169 radiomics features was standardized, which enabled verification and calibration of different radiomics software. © RSNA, 2020 Online supplemental material is available for this article. See also the editorial by Kuhl and Truhn in this issue.


Assuntos
Biomarcadores/análise , Processamento de Imagem Assistida por Computador/normas , Software , Calibragem , Fluordesoxiglucose F18 , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Imageamento por Ressonância Magnética , Imagens de Fantasmas , Fenótipo , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Reprodutibilidade dos Testes , Sarcoma/diagnóstico por imagem , Tomografia Computadorizada por Raios X
9.
Mol Imaging Biol ; 22(4): 1132-1148, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32185618

RESUMO

PURPOSE: Considerable progress has been made in the assessment and management of non-small cell lung cancer (NSCLC) patients based on mutation status in the epidermal growth factor receptor (EGFR) and Kirsten rat sarcoma viral oncogene (KRAS). At the same time, NSCLC management through KRAS and EGFR mutation profiling faces challenges. In the present work, we aimed to evaluate a comprehensive radiomics framework that enabled prediction of EGFR and KRAS mutation status in NSCLC patients based on radiomic features from low-dose computed tomography (CT), contrast-enhanced diagnostic quality CT (CTD), and positron emission tomography (PET) imaging modalities and use of machine learning algorithms. METHODS: Our study involved NSCLC patients including 150 PET, low-dose CT, and CTD images. Radiomic features from original and preprocessed (including 64 bin discretizing, Laplacian-of-Gaussian (LOG), and Wavelet) images were extracted. Conventional clinically used standard uptake value (SUV) parameters and metabolic tumor volume (MTV) were also obtained from PET images. Highly correlated features were pre-eliminated, and false discovery rate (FDR) correction was performed with the resulting q-values reported for univariate analysis. Six feature selection methods and 12 classifiers were then used for multivariate prediction of gene mutation status (provided by polymerase chain reaction (PCR)) in patients. We performed 10-fold cross-validation for model tuning to improve robustness, and our developed models were assessed on an independent validation set with 68 patients (common in all three imaging modalities). The average area under the receiver operator characteristic curve (AUC) was utilized for performance evaluation. RESULTS: The best predictive power for conventional PET parameters was achieved by SUVpeak (AUC 0.69, p value = 0.0002) and MTV (AUC 0.55, p value = 0.0011) for EGFR and KRAS, respectively. Univariate analysis of extracted radiomics features improved AUC performance to 0.75 (q-value 0.003, Short-Run Emphasis feature of GLRLM from LOG preprocessed image of PET with sigma value 1.5) and 0.71 (q-value 0.00005, Large Dependence Low Gray-Level Emphasis feature of GLDM in LOG preprocessed image of CTD with sigma value 5) for EGFR and KRAS, respectively. Furthermore, multivariate machine learning-based AUC performances were significantly improved to 0.82 for EGFR (LOG preprocessed image of PET with sigma 3 with variance threshold (VT) feature selector and stochastic gradient descent (SGD) classifier (q-value = 4.86E-05) and 0.83 for KRAS (LOG preprocessed image of CT with sigma 3.5 with select model (SM) feature selector and SGD classifier (q-value = 2.81E-09). CONCLUSION: Our work demonstrated that non-invasive and reliable radiomics analysis can be successfully used to predict EGFR and KRAS mutation status in NSCLC patients. We demonstrated that radiomic features extracted from different image-feature sets could be used for EGFR and KRAS mutation status prediction in NSCLC patients and showed improved predictive power relative to conventional image-derived metrics.


Assuntos
Algoritmos , Carcinoma Pulmonar de Células não Pequenas/genética , Receptores ErbB/genética , Sequenciamento de Nucleotídeos em Larga Escala , Aprendizado de Máquina , Imagem Multimodal , Mutação/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Idoso , Área Sob a Curva , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Feminino , Genômica , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/genética , Masculino , Análise Multivariada , Tomografia por Emissão de Pósitrons
10.
Mol Imaging Biol ; 22(3): 730-738, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31338709

RESUMO

PURPOSE: To identify optimal machine learning methods for radiomics-based differentiation of local recurrence versus inflammation from post-treatment nasopharyngeal positron emission tomography/X-ray computed tomography (PET/CT) images. PROCEDURES: Seventy-six nasopharyngeal carcinoma (NPC) patients were enrolled (41/35 local recurrence/inflammation as confirmed by pathology). Four hundred eighty-seven radiomics features were extracted from PET images for each patient. The diagnostic performance was investigated for 42 cross-combinations derived from 6 feature selection methods and 7 classifiers. Of the original cohort, 70 % was applied for feature selection and classifier development, and the remaining 30 % used as an independent validation set. The diagnostic performance was evaluated using area under the ROC curve (AUC), test error, sensitivity, and specificity. Furthermore, the performance of the radiomics signatures against routine features was statistically compared using DeLong's method. RESULTS: The cross-combination fisher score (FSCR) + k-nearest neighborhood (KNN), FSCR + support vector machines with radial basis function kernel (RBF-SVM), FSCR + random forest (RF), and minimum redundancy maximum relevance (MRMR) + RBF-SVM outperformed others in terms of accuracy (AUC 0.883, 0.867, 0.892, 0.883; sensitivity 0.833, 0.864, 0.831, 0.750; specificity 1, 1, 0.873, 1) and reliability (test error 0.091, 0.136, 0.150, 0.136). Compared with conventional metrics, the radiomics signatures showed higher AUC values (0.867-0.892 vs. 0.817), though the differences were not statistically significant (p = 0.462-0.560). CONCLUSION: This study identified the most accurate and reliable machine learning methods, which could enhance the application of radiomics methods in the precision of diagnosis of NPC.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Aprendizado de Máquina , Carcinoma Nasofaríngeo/diagnóstico por imagem , Neoplasias Nasofaríngeas/diagnóstico por imagem , Recidiva Local de Neoplasia/diagnóstico por imagem , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Algoritmos , Diagnóstico Diferencial , Feminino , Humanos , Inflamação/diagnóstico por imagem , Inflamação/patologia , Inflamação/terapia , Masculino , Carcinoma Nasofaríngeo/patologia , Carcinoma Nasofaríngeo/terapia , Neoplasias Nasofaríngeas/patologia , Neoplasias Nasofaríngeas/terapia , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/terapia , Curva ROC , Estudos Retrospectivos
11.
IEEE J Biomed Health Inform ; 24(8): 2268-2277, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31804945

RESUMO

To characterize intra-tumor heterogeneity comprehensively, we propose a multi-level fusion strategy to combine PET and CT information at the image-, matrix-and feature-levels towards improved prognosis. Specifically, we developed fusion radiomics in the context of 3 prognostic outcomes in a multi-center setting (4 centers) involving 296 head & neck cancer patients. Eight clinical parameters were first utilized to build a (1) clinical model. We also built models by extracting 127 radiomics features from (2) PET images alone; (3-8) PET and CT images fused via wavelet-based fusion (WF) using CT-weights of 0.2, 0.4, 0.6 and 0.8, gradient transfer fusion (GTF), and guided filtering-based fusion (GFF); (9) fused matrices (sumMat); (10-11) fused features constructed via feature averaging (avgFea) and feature concatenation (conFea); and finally, (12) CT images alone; above models were also expanded to include both clinical and radiomics features. Seven variations of training and testing partitions were investigated. Highest performance in 5, 6 and 5 partitions was achieved by image-level fusion strategies for RFS, MFS and OS prediction, respectively. Among all partitions, WF0.6 and WF0.8 showed significantly higher performance than CT model for RFS (C-index: 0.60 ± 0.04 vs. 0.56 ± 0.03, p-value: 0.015) and MFS (C-index: 0.71 ± 0.13 vs. 0.62 ± 0.08, p-value: 0.020) predictions, respectively. In partition CER 23 vs. 14, WF0.6 significantly outperformed Clinical model for RFS prediction (C-index: 0.67 vs. 0.53, p-value: 0.003); both avgFea and WF0.6 showed C-index of 0.64 and significantly higher than that of PET only (C-index: 0.51, p-value: 0.018 and 0.031, respectively) for OS prediction. Fusion radiomics modeling showed varying improvements compared to single modality models for different outcome predictions in different partitions, highlighting the importance of generalizing radiomics models. Image-level fusion holds potential to capture more useful characteristics.


Assuntos
Neoplasias de Cabeça e Pescoço/diagnóstico por imagem , Interpretação de Imagem Assistida por Computador/métodos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Cabeça/diagnóstico por imagem , Humanos , Masculino , Pessoa de Meia-Idade , Pescoço/diagnóstico por imagem , Prognóstico , Adulto Jovem
12.
Eur J Radiol ; 113: 101-109, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30927933

RESUMO

OBJECTIVE: We aimed to improve prediction of outcome for patients with colorectal liver metastases, via prognostic models incorporating PET-derived measures, including radiomic features that move beyond conventional standard uptake value (SUV) measures. PATIENTS AND METHODS: A range of parameters including volumetric and heterogeneity measures were derived from FDG PET images of 52 patients with colorectal intrahepatic-only metastases (29 males and 23 females; mean age 62.9 years [SD 9.8; range 32-82]). The patients underwent PET/CT imaging as part of the clinical workup prior to final decision on treatment. Univariate and multivariate models were implemented, which included statistical considerations (to discourage false discovery and overfitting), to predict overall survival (OS), progression-free survival (PFS) and event-free survival (EFS). Kaplan-Meier survival analyses were performed, where the subjects were divided into high-risk and low-risk groups, from which the hazard ratios (HR) were computed via Cox proportional hazards regression. RESULTS: Commonly-invoked SUV metrics performed relatively poorly for different prediction tasks (SUVmax HR = 1.48, 0.83 and 1.16; SUVpeak HR = 2.05, 1.93, and 1.64, for OS, PFS and EFS, respectively). By contrast, the number of liver metastases and metabolic tumor volume (MTV) each performed well (with respective HR values of 2.71, 2.61 and 2.42, and 2.62, 1.96 and 2.29, for OS, PFS and EFS). Total lesion glycolysis (TLG) also resulted in similar performance as MTV. Multivariate prognostic modeling incorporating different features (including those quantifying intra-tumor heterogeneity) resulted in further enhanced prediction. Specifically, HR values of 4.29, 4.02 and 3.20 (p-values = 0.00004, 0.0019 and 0.0002) were obtained for OS, PFS and EFS, respectively. CONCLUSIONS: PET-derived measures beyond commonly invoked SUV parameters hold significant potential towards improved prediction of clinical outcome in patients with liver metastases, especially when utilizing multivariate models.


Assuntos
Neoplasias Colorretais , Neoplasias Hepáticas/diagnóstico por imagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Intervalo Livre de Doença , Feminino , Fluordesoxiglucose F18/metabolismo , Glicólise/fisiologia , Humanos , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/secundário , Masculino , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Tomografia por Emissão de Pósitrons , Prognóstico , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Carga Tumoral , Adulto Jovem
13.
J Med Imaging (Bellingham) ; 4(1): 011011, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28331883

RESUMO

Recently, a class of no-gold-standard (NGS) techniques have been proposed to evaluate quantitative imaging methods using patient data. These techniques provide figures of merit (FoMs) quantifying the precision of the estimated quantitative value without requiring repeated measurements and without requiring a gold standard. However, applying these techniques to patient data presents several practical difficulties including assessing the underlying assumptions, accounting for patient-sampling-related uncertainty, and assessing the reliability of the estimated FoMs. To address these issues, we propose statistical tests that provide confidence in the underlying assumptions and in the reliability of the estimated FoMs. Furthermore, the NGS technique is integrated within a bootstrap-based methodology to account for patient-sampling-related uncertainty. The developed NGS framework was applied to evaluate four methods for segmenting lesions from F-Fluoro-2-deoxyglucose positron emission tomography images of patients with head-and-neck cancer on the task of precisely measuring the metabolic tumor volume. The NGS technique consistently predicted the same segmentation method as the most precise method. The proposed framework provided confidence in these results, even when gold-standard data were not available. The bootstrap-based methodology indicated improved performance of the NGS technique with larger numbers of patient studies, as was expected, and yielded consistent results as long as data from more than 80 lesions were available for the analysis.

14.
Phys Med Biol ; 62(12): 5149-5179, 2017 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-28338471

RESUMO

Point-spread function (PSF) modeling offers the ability to account for resolution degrading phenomena within the PET image generation framework. PSF modeling improves resolution and enhances contrast, but at the same time significantly alters image noise properties and induces edge overshoot effect. Thus, studying the effect of PSF modeling on quantitation task performance can be very important. Frameworks explored in the past involved a dichotomy of PSF versus no-PSF modeling. By contrast, the present work focuses on quantitative performance evaluation of standard uptake value (SUV) PET images, while incorporating a wide spectrum of PSF models, including those that under- and over-estimate the true PSF, for the potential of enhanced quantitation of SUVs. The developed framework first analytically models the true PSF, considering a range of resolution degradation phenomena (including photon non-collinearity, inter-crystal penetration and scattering) as present in data acquisitions with modern commercial PET systems. In the context of oncologic liver FDG PET imaging, we generated 200 noisy datasets per image-set (with clinically realistic noise levels) using an XCAT anthropomorphic phantom with liver tumours of varying sizes. These were subsequently reconstructed using the OS-EM algorithm with varying PSF modelled kernels. We focused on quantitation of both SUVmean and SUVmax, including assessment of contrast recovery coefficients, as well as noise-bias characteristics (including both image roughness and coefficient of-variability), for different tumours/iterations/PSF kernels. It was observed that overestimated PSF yielded more accurate contrast recovery for a range of tumours, and typically improved quantitative performance. For a clinically reasonable number of iterations, edge enhancement due to PSF modeling (especially due to over-estimated PSF) was in fact seen to lower SUVmean bias in small tumours. Overall, the results indicate that exactly matched PSF modeling does not offer optimized PET quantitation, and that PSF overestimation may provide enhanced SUV quantitation. Furthermore, generalized PSF modeling may provide a valuable approach for quantitative tasks such as treatment-response assessment and prognostication.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Modelos Teóricos , Tomografia por Emissão de Pósitrons , Algoritmos , Humanos , Neoplasias Hepáticas/diagnóstico por imagem , Imagens de Fantasmas , Razão Sinal-Ruído
15.
Phys Med Biol ; 61(1): 227-42, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26639024

RESUMO

Oncologic PET images provide valuable information that can enable enhanced prognosis of disease. Nonetheless, such information is simplified significantly in routine clinical assessment to meet workflow constraints. Examples of typical FDG PET metrics include: (i) SUVmax, (2) total lesion glycolysis (TLG), and (3) metabolic tumor volume (MTV). We have derived and implemented a novel metric for tumor quantification, inspired in essence by a model of generalized equivalent uniform dose as used in radiation therapy. The proposed metric, denoted generalized effective total uptake (gETU), is attractive as it encompasses the abovementioned commonly invoked metrics, and generalizes them, for both homogeneous and heterogeneous tumors, using a single parameter a. We evaluated this new metric for improved overall survival (OS) prediction on two different baseline FDG PET/CT datasets: (a) 113 patients with squamous cell cancer of the oropharynx, and (b) 72 patients with locally advanced pancreatic adenocarcinoma. Kaplan-Meier survival analysis was performed, where the subjects were subdivided into two groups using the median threshold, from which the hazard ratios (HR) were computed in Cox proportional hazards regression. For the oropharyngeal cancer dataset, MTV, TLG, SUVmax, SUVmean and SUVpeak produced HR values of 1.86, 3.02, 1.34, 1.36 and 1.62, while the proposed gETU metric for a = 0.25 (greater emphasis on volume information) enabled significantly enhanced OS prediction with HR = 3.94. For the pancreatic cancer dataset, MTV, TLG, SUVmax, SUVmean and SUVpeak resulted in HR values of 1.05, 1.25, 1.42, 1.45 and 1.52, while gETU at a = 3.2 (greater emphasis on SUV information) arrived at an improved HR value of 1.61. Overall, the proposed methodology allows placement of differing degrees of emphasis on tumor volume versus uptake for different types of tumors to enable enhanced clinical outcome prediction.


Assuntos
Adenocarcinoma/diagnóstico por imagem , Carcinoma de Células Escamosas/diagnóstico por imagem , Imagem Multimodal/métodos , Neoplasias Orofaríngeas/diagnóstico por imagem , Neoplasias Pancreáticas/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Tomografia Computadorizada por Raios X/métodos , Adulto , Idoso , Feminino , Fluordesoxiglucose F18/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Compostos Radiofarmacêuticos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...