80 research outputs found

    Semi-automated 18F-FDG PET segmentation methods for tumor volume determination in Non-Hodgkin lymphoma patients:a literature review, implementation and multi-threshold evaluation

    Get PDF
    In the treatment of Non-Hodgkin lymphoma (NHL), multiple therapeutic options are available. Improving outcome predictions are essential to optimize treatment. The metabolic active tumor volume (MATV) has shown to be a prognostic factor in NHL. It is usually retrieved using semi-automated thresholding methods based on standardized uptake values (SUV), calculated from 18F-Fluorodeoxyglucose Positron Emission Tomography (18F-FDG PET) images. However, there is currently no consensus method for NHL. The aim of this study was to review literature on different segmentation methods used, and to evaluate selected methods by using an in house created software tool. A software tool, MUltiple SUV Threshold (MUST)-segmenter was developed where tumor locations are identified by placing seed-points on the PET images, followed by subsequent region growing. Based on a literature review, 9 SUV thresholding methods were selected and MATVs were extracted. The MUST-segmenter was utilized in a cohort of 68 patients with NHL. Differences in MATVs were assessed with paired t-tests, and correlations and distributions figures. High variability and significant differences between the MATVs based on different segmentation methods (p < 0.05) were observed in the NHL patients. Median MATVs ranged from 35 to 211 cc. No consensus for determining MATV is available based on the literature. Using the MUST-segmenter with 9 selected SUV thresholding methods, we demonstrated a large and significant variation in MATVs. Identifying the most optimal segmentation method for patients with NHL is essential to further improve predictions of toxicity, response, and treatment outcomes, which can be facilitated by the MUST-segmenter

    Comparison of computed tomography image features extracted by radiomics, self-supervised learning and end-to-end deep learning for outcome prediction of oropharyngeal cancer

    Get PDF
    Background and purpose: To compare the prediction performance of image features of computed tomography (CT) images extracted by radiomics, self-supervised learning and end-to-end deep learning for local control (LC), regional control (RC), locoregional control (LRC), distant metastasis-free survival (DMFS), tumor-specific survival (TSS), overall survival (OS) and disease-free survival (DFS) of oropharyngeal squamous cell carcinoma (OPSCC) patients after (chemo)radiotherapy.Methods and materials: The OPC-Radiomics dataset was used for model development and independent internal testing and the UMCG-OPC set for external testing. Image features were extracted from the Gross Tumor Volume contours of the primary tumor (GTVt) regions in CT scans when using radiomics or a self-supervised learning-based method (autoencoder). Clinical and combined (radiomics, autoencoder or end-to-end) models were built using multivariable Cox proportional-hazard analysis with clinical features only and both clinical and image features for LC, RC, LRC, DMFS, TSS, OS and DFS prediction, respectively.Results: In the internal test set, combined autoencoder models performed better than clinical models and combined radiomics models for LC, RC, LRC, DMFS, TSS and DFS prediction (largest improvements in C-index: 0.91 vs. 0.76 in RC and 0.74 vs. 0.60 in DMFS). In the external test set, combined radiomics models performed better than clinical and combined autoencoder models for all endpoints (largest improvements in LC, 0.82 vs. 0.71). Furthermore, combined models performed better in risk stratification than clinical models and showed good calibration for most endpoints.Conclusions: Image features extracted using self-supervised learning showed best internal prediction performance while radiomics features have better external generalizability.</p

    Can we safely reduce the radiation dose to the heart while compromising the dose to the lungs in oesophageal cancer patients?

    Get PDF
    PURPOSE: The aim of this study was to evaluate which clinical and treatment-related factors are associated with heart and lung toxicity in oesophageal cancer patients treated with chemoradiation (CRT). The secondary objective was to analyse whether these toxicities are associated with overall survival (OS) MATERIALS AND METHODS: The study population consisted of a retrospective cohort of 216 oesophageal cancer patients treated with curative CRT. Clinical and treatment related factors were analysed for OS and new pulmonary and cardiac events by multivariable regression analyses. The effect of these toxicities on OS was assessed by Kaplan Meyer analyses. RESULTS: Multivariable analysis revealed that pulmonary toxicity was best predicted by the mean lung dose. Cardiac complications were diverse; the most frequently occurring complication was pericardial effusion. Several cardiac dose parameters correlated with this endpoint. Patients developing radiation pneumonitis had significantly worse OS than patients without radiation pneumonitis, while no difference was observed in OS between patients with and without pericardial effusion. OS was best predicted by the V45 of the lung and tumour stage. None of the cardiac dose parameters predicted OS in multivariable analyses. CONCLUSION: Cardiac dose volume parameters predicted the risk of pericardial effusion and pulmonary dose volume parameters predicted the risk of radiation pneumonitis. However, in this patient cohort, pulmonary DVH parameters (V45) were more important for OS than cardiac DVH parameters. These results suggest that reducing the cardiac dose at the expense of the dose to the lungs might not always be a good strategy in oesophageal cancer patients

    Development of a prediction model for late urinary incontinence, hematuria, pain and voiding frequency among irradiated prostate cancer patients

    Get PDF
    BACKGROUND AND PURPOSE:Incontinence, hematuria, voiding frequency and pain during voiding are possible side effects of radiotherapy among patients treated for prostate cancer. The objective of this study was to develop multivariable NTCP models for these side effects. MATERIAL AND METHODS:This prospective cohort study was composed of 243 patients with localized or locally advanced prostate cancer (stage T1-3). Genito-urinary (GU) toxicity was assessed using a standardized follow-up program. The GU toxicity endpoints were scored using the Common Terminology Criteria for Adverse Events version 3.0 (CTCAE 3.0) scoring system. The full bladder and different anatomical subregions within the bladder were delineated. A least absolute shrinkage and selection operator (LASSO) logistic regression analysis was used to analyze dose volume effects on the four individual endpoints. RESULTS:In the univariable analysis, urinary incontinence was significantly associated with dose distributions in the trigone (V55-V75, mean). Hematuria was significantly associated with the bladder wall dose (V40-V75, mean), bladder dose (V70-V75), cardiovascular disease and anticoagulants use. Pain during urinating was associated with the dose to the trigone (V50-V75, mean) and with trans transurethral resection of the prostate (TURP). In the final multivariable model urinary incontinence was associated with the mean dose of the trigone. Hematuria was associated with bladder wall dose (V75) and cardiovascular disease, while pain during urinating was associated with trigone dose (V75) and TURP. No significant associations were found for increase in voiding frequency. CONCLUSIONS:Radiation-induced urinary side effects are associated with dose distributions to different organs as risk. Given the dose effect relationships found, decreasing the dose to the trigone and bladder wall may reduce the incidence of incontinence, pain during voiding and hematuria, respectively

    Head and Neck Cancer Primary Tumor Auto Segmentation Using Model Ensembling of Deep Learning in PET/CT Images

    Get PDF
    Auto-segmentation of primary tumors in oropharyngeal cancer using PET/CT images is an unmet need that has the potential to improve radiation oncology workflows. In this study, we develop a series of deep learning models based on a 3D Residual Unet (ResUnet) architecture that can segment oropharyngeal tumors with high performance as demonstrated through internal and external validation of large-scale datasets (training size = 224 patients, testing size = 101 patients) as part of the 2021 HECKTOR Challenge. Specifically, we leverage ResUNet models with either 256 or 512 bottleneck layer channels that demonstrate internal validation (10-fold cross-validation) mean Dice similarity coefficient (DSC) up to 0.771 and median 95% Hausdorff distance (95% HD) as low as 2.919 mm. We employ label fusion ensemble approaches, including Simultaneous Truth and Performance Level Estimation (STAPLE) and a voxel-level threshold approach based on majority voting (AVERAGE), to generate consensus segmentations on the test data by combining the segmentations produced through different trained cross-validation models. We demonstrate that our best performing ensembling approach (256 channels AVERAGE) achieves a mean DSC of 0.770 and median 95% HD of 3.143 mm through independent external validation on the test set. Our DSC and 95% HD test results are within 0.01 and 0.06 mm of the top ranked model in the competition, respectively. Concordance of internal and external validation results suggests our models are robust and can generalize well to unseen PET/CT data. We advocate that ResUNet models coupled to label fusion ensembling approaches are promising candidates for PET/CT oropharyngeal primary tumors auto-segmentation. Future investigations should target the ideal combination of channel combinations and label fusion strategies to maximize segmentation performance.</p

    Improving automatic delineation for head and neck organs at risk by Deep Learning Contouring

    Get PDF
    INTRODUCTION: Adequate head and neck (HN) organ-at-risk (OAR) delineation is crucial for HN radiotherapy and for investigating the relationships between radiation dose to OARs and radiation-induced side effects. The automatic contouring algorithms that are currently in clinical use, such as atlas-based contouring (ABAS), leave room for improvement. The aim of this study was to use a comprehensive evaluation methodology to investigate the performance of HN OAR auto-contouring when using deep learning contouring (DLC), compared to ABAS. METHODS: The DLC neural network was trained on 589 HN cancer patients. DLC was compared to ABAS by providing each method with an independent validation cohort of 104 patients, which had also been manually contoured. For each of the 22 OAR contours - glandular, upper digestive tract and central nervous system (CNS)-related structures - the dice similarity coefficient (DICE), and absolute mean and max dose differences (|Δmean-dose| and |Δmax-dose|) performance measures were obtained. For a subset of 7 OARs, an evaluation of contouring time, inter-observer variation and subjective judgement was performed. RESULTS: DLC resulted in equal or significantly improved quantitative performance measures in 19 out of 22 OARs, compared to the ABAS (DICE/|Δmean dose|/|Δmax dose|: 0.59/4.2/4.1 Gy (ABAS); 0.74/1.1/0.8 Gy (DLC)). The improvements were mainly for the glandular and upper digestive tract OARs. DLC significantly reduced the delineation time for the inexperienced observer. The subjective evaluation showed that DLC contours were more often preferable to the ABAS contours overall, were considered to be more precise, and more often confused with manual contours. Manual contours still outperformed both DLC and ABAS; however, DLC results were within or bordering the inter-observer variability for the manual edited contours in this cohort. CONCLUSION: The DLC, trained on a large HN cancer patient cohort, outperformed the ABAS for the majority of HN OARs

    (18)F-FDG PET image biomarkers improve prediction of late radiation-induced xerostomia

    Get PDF
    BACKGROUND AND PURPOSE: Current prediction of radiation-induced xerostomia 12months after radiotherapy (Xer12m) is based on mean parotid gland dose and baseline xerostomia (Xerbaseline) scores. The hypothesis of this study was that prediction of Xer12m is improved with patient-specific characteristics extracted from (18)F-FDG PET images, quantified in PET image biomarkers (PET-IBMs). PATIENTS AND METHODS: Intensity and textural PET-IBMs of the parotid gland were collected from pre-treatment (18)F-FDG PET images of 161 head and neck cancer patients. Patient-rated toxicity was prospectively collected. Multivariable logistic regression models resulting from step-wise forward selection and Lasso regularisation were internally validated by bootstrapping. The reference model with parotid gland dose and Xerbaseline was compared with the resulting PET-IBM models. RESULTS: High values of the intensity PET-IBM (90th percentile (P90)) and textural PET-IBM (Long Run High Grey-level Emphasis 3 (LRHG3E)) were significantly associated with lower risk of Xer12m. Both PET-IBMs significantly added in the prediction of Xer12m to the reference model. The AUC increased from 0.73 (0.65-0.81) (reference model) to 0.77 (0.70-0.84) (P90) and 0.77 (0.69-0.84) (LRHG3E). CONCLUSION: Prediction of Xer12m was significantly improved with pre-treatment PET-IBMs, indicating that high metabolic parotid gland activity is associated with lower risk of developing late xerostomia. This study highlights the potential of incorporating patient-specific PET-derived functional characteristics into NTCP model development

    Multi-organ spatial stratification of 3-D dose distributions improves risk prediction of long-term self-reported severe symptoms in oropharyngeal cancer patients receiving radiotherapy:development of a pre-treatment decision support tool

    Get PDF
    PURPOSE: Identify Oropharyngeal cancer (OPC) patients at high-risk of developing long-term severe radiation-associated symptoms using dose volume histograms for organs-at-risk, via unsupervised clustering.MATERIAL AND METHODS: All patients were treated using radiation therapy for OPC. Dose-volume histograms of organs-at-risk were extracted from patients' treatment plans. Symptom ratings were collected via the MD Anderson Symptom Inventory (MDASI) given weekly during, and 6 months post-treatment. Drymouth, trouble swallowing, mucus, and vocal dysfunction were selected for analysis in this study. Patient stratifications were obtained by applying Bayesian Mixture Models with three components to patient's dose histograms for relevant organs. The clusters with the highest total mean doses were translated into dose thresholds using rule mining. Patient stratifications were compared against Tumor staging information using multivariate likelihood ratio tests. Model performance for prediction of moderate/severe symptoms at 6 months was compared against normal tissue complication probability (NTCP) models using cross-validation.RESULTS: A total of 349 patients were included for long-term symptom prediction. High-risk clusters were significantly correlated with outcomes for severe late drymouth (p &lt;.0001, OR = 2.94), swallow (p = .002, OR = 5.13), mucus (p = .001, OR = 3.18), and voice (p = .009, OR = 8.99). Simplified clusters were also correlated with late severe symptoms for drymouth (p &lt;.001, OR = 2.77), swallow (p = .01, OR = 3.63), mucus (p = .01, OR = 2.37), and voice (p &lt;.001, OR = 19.75). Proposed cluster stratifications show better performance than NTCP models for severe drymouth (AUC.598 vs.559, MCC.143 vs.062), swallow (AUC.631 vs.561, MCC.20 vs -.030), mucus (AUC.596 vs.492, MCC.164 vs -.041), and voice (AUC.681 vs.555, MCC.181 vs -.019). Simplified dose thresholds also show better performance than baseline models for predicting late severe ratings for all symptoms.CONCLUSION: Our results show that leveraging the 3-D dose histograms from radiation therapy plan improves stratification of patients according to their risk of experiencing long-term severe radiation associated symptoms, beyond existing NTPC models. Our rule-based method can approximate our stratifications with minimal loss of accuracy and can proactively identify risk factors for radiation-associated toxicity.</p
    • …
    corecore