Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
PLoS One ; 18(7): e0288695, 2023.
Article En | MEDLINE | ID: mdl-37471412

Experiencing food craving is nearly ubiquitous and has several negative pathological impacts prompting an increase in recent craving-related research. Food cue-reactivity tasks are often used to study craving, but most paradigms ignore the individual food preferences of participants, which could confound the findings. We explored the neuropsychological correlates of food craving preference using psychophysical tasks on human participants considering their individual food preferences in a multisensory food exposure set-up. Participants were grouped into Liked Food Exposure (LFE), Disliked Food Exposure (DFE), and Neutral Control (NEC) based on their preference for sweet and savory items. Participants reported their momentary craving for the displayed food stimuli through the desire scale and bidding scale (willingness to pay) pre and post multisensory exposure. Participants were exposed to food items they either liked or disliked. Our results asserted the effect of the multisensory food exposure showing a statistically significant increase in food craving for DFE participants postexposure to disliked food items. Using computational models and statistical methods, we also show that the desire for food does not necessarily translate to a willingness to pay every time, and instantaneous subjective valuation of food craving is an important parameter for subsequent action. Our results further demonstrate the role of parietal N200 and centro-parietal P300 in reversing food preference and possibly point to the decrease of inhibitory control in up-regulating craving for disliked food.


Cues , Food Preferences , Humans , Food Preferences/psychology , Craving/physiology , Food , Emotions
2.
Nucl Med Commun ; 44(8): 709-718, 2023 08 01.
Article En | MEDLINE | ID: mdl-37132272

OBJECTIVES: Detection of residual oesophageal cancer after neoadjuvant chemoradiotherapy (nCRT) is important to guide treatment decisions regarding standard oesophagectomy or active surveillance. The aim was to validate previously developed 18 F-FDG PET-based radiomic models to detect residual local tumour and to repeat model development (i.e. 'model extension') in case of poor generalisability. METHODS: This was a retrospective cohort study in patients collected from a prospective multicentre study in four Dutch institutes. Patients underwent nCRT followed by oesophagectomy between 2013 and 2019. Outcome was tumour regression grade (TRG) 1 (0% tumour) versus TRG 2-3-4 (≥1% tumour). Scans were acquired according to standardised protocols. Discrimination and calibration were assessed for the published models with optimism-corrected AUCs >0.77. For model extension, the development and external validation cohorts were combined. RESULTS: Baseline characteristics of the 189 patients included [median age 66 years (interquartile range 60-71), 158/189 male (84%), 40/189 TRG 1 (21%) and 149/189 (79%) TRG 2-3-4] were comparable to the development cohort. The model including cT stage plus the feature 'sum entropy' had best discriminative performance in external validation (AUC 0.64, 95% confidence interval 0.55-0.73), with a calibration slope and intercept of 0.16 and 0.48 respectively. An extended bootstrapped LASSO model yielded an AUC of 0.65 for TRG 2-3-4 detection. CONCLUSION: The high predictive performance of the published radiomic models could not be replicated. The extended model had moderate discriminative ability. The investigated radiomic models appeared inaccurate to detect local residual oesophageal tumour and cannot be used as an adjunct tool for clinical decision-making in patients.


Esophageal Neoplasms , Fluorodeoxyglucose F18 , Humans , Male , Aged , Retrospective Studies , Neoadjuvant Therapy/methods , Prospective Studies , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/therapy , Esophageal Neoplasms/pathology , Chemoradiotherapy
3.
J Pathol Inform ; 14: 100192, 2023.
Article En | MEDLINE | ID: mdl-36818020

Treatment of patients with oesophageal and gastric cancer (OeGC) is guided by disease stage, patient performance status and preferences. Lymph node (LN) status is one of the strongest prognostic factors for OeGC patients. However, survival varies between patients with the same disease stage and LN status. We recently showed that LN size from patients with OeGC might also have prognostic value, thus making delineations of LNs essential for size estimation and the extraction of other imaging biomarkers. We hypothesized that a machine learning workflow is able to: (1) find digital H&E stained slides containing LNs, (2) create a scoring system providing degrees of certainty for the results, and (3) delineate LNs in those images. To train and validate the pipeline, we used 1695 H&E slides from the OE02 trial. The dataset was divided into training (80%) and validation (20%). The model was tested on an external dataset of 826 H&E slides from the OE05 trial. U-Net architecture was used to generate prediction maps from which predefined features were extracted. These features were subsequently used to train an XGBoost model to determine if a region truly contained a LN. With our innovative method, the balanced accuracies of the LN detection were 0.93 on the validation dataset (0.83 on the test dataset) compared to 0.81 (0.81) on the validation (test) datasets when using the standard method of thresholding U-Net predictions to arrive at a binary mask. Our method allowed for the creation of an "uncertain" category, and partly limited false-positive predictions on the external dataset. The mean Dice score was 0.73 (0.60) per-image and 0.66 (0.48) per-LN for the validation (test) datasets. Our pipeline detects images with LNs more accurately than conventional methods, and high-throughput delineation of LNs can facilitate future LN content analyses of large datasets.

4.
Front Digit Health ; 5: 1303261, 2023.
Article En | MEDLINE | ID: mdl-38586126

The aim of this study was to develop and evaluate a proof-of-concept open-source individualized Patient Decision Aid (iPDA) with a group of patients, physicians, and computer scientists. The iPDA was developed based on the International Patient Decision Aid Standards (IPDAS). A previously published questionnaire was adapted and used to test the user-friendliness and content of the iPDA. The questionnaire contained 40 multiple-choice questions, and answers were given on a 5-point Likert Scale (1-5) ranging from "strongly disagree" to "strongly agree." In addition to the questionnaire, semi-structured interviews were conducted with patients. We performed a descriptive analysis of the responses. The iPDA was evaluated by 28 computer scientists, 21 physicians, and 13 patients. The results demonstrate that the iPDA was found valuable by 92% (patients), 96% (computer scientists), and 86% (physicians), while the treatment information was judged useful by 92%, 96%, and 95%, respectively. Additionally, the tool was thought to be motivating for patients to actively engage in their treatment by 92%, 93%, and 91% of the above respondents groups. More multimedia components and less text were suggested by the respondents as ways to improve the tool and user interface. In conclusion, we successfully developed and tested an iPDA for patients with stage I-II Non-Small Cell Lung Cancer (NSCLC).

5.
Biomedicines ; 10(11)2022 Oct 23.
Article En | MEDLINE | ID: mdl-36359199

(1) Background: The main aim was to develop a prototype application that would serve as an open-source repository for a curated subset of predictive and prognostic models regarding oncology, and provide a user-friendly interface for the included models to allow online calculation. The focus of the application is on providing physicians and health professionals with patient-specific information regarding treatment plans, survival rates, and side effects for different expected treatments. (2) Methods: The primarily used models were the ones developed by our research group in the past. This selection was completed by a number of models, addressing the same cancer types but focusing on other outcomes that were selected based on a literature search in PubMed and Medline databases. All selected models were publicly available and had been validated TRIPOD (Transparent Reporting of studies on prediction models for Individual Prognosis Or Diagnosis) type 3 or 2b. (3) Results: The open source repository currently incorporates 18 models from different research groups, evaluated on datasets from different countries. Model types included logistic regression, Cox regression, and recursive partition analysis (decision trees). (4) Conclusions: An application was developed to enable physicians to complement their clinical judgment with user-friendly patient-specific predictions using models that have received internal/external validation. Additionally, this platform enables researchers to display their work, enhancing the use and exposure of their models.

6.
Front Oncol ; 12: 920393, 2022.
Article En | MEDLINE | ID: mdl-35912214

Introduction: There is a cumulative risk of 20-40% of developing brain metastases (BM) in solid cancers. Stereotactic radiotherapy (SRT) enables the application of high focal doses of radiation to a volume and is often used for BM treatment. However, SRT can cause adverse radiation effects (ARE), such as radiation necrosis, which sometimes cause irreversible damage to the brain. It is therefore of clinical interest to identify patients at a high risk of developing ARE. We hypothesized that models trained with radiomics features, deep learning (DL) features, and patient characteristics or their combination can predict ARE risk in patients with BM before SRT. Methods: Gadolinium-enhanced T1-weighted MRIs and characteristics from patients treated with SRT for BM were collected for a training and testing cohort (N = 1,404) and a validation cohort (N = 237) from a separate institute. From each lesion in the training set, radiomics features were extracted and used to train an extreme gradient boosting (XGBoost) model. A DL model was trained on the same cohort to make a separate prediction and to extract the last layer of features. Different models using XGBoost were built using only radiomics features, DL features, and patient characteristics or a combination of them. Evaluation was performed using the area under the curve (AUC) of the receiver operating characteristic curve on the external dataset. Predictions for individual lesions and per patient developing ARE were investigated. Results: The best-performing XGBoost model on a lesion level was trained on a combination of radiomics features and DL features (AUC of 0.71 and recall of 0.80). On a patient level, a combination of radiomics features, DL features, and patient characteristics obtained the best performance (AUC of 0.72 and recall of 0.84). The DL model achieved an AUC of 0.64 and recall of 0.85 per lesion and an AUC of 0.70 and recall of 0.60 per patient. Conclusion: Machine learning models built on radiomics features and DL features extracted from BM combined with patient characteristics show potential to predict ARE at the patient and lesion levels. These models could be used in clinical decision making, informing patients on their risk of ARE and allowing physicians to opt for different therapies.

7.
Ther Adv Med Oncol ; 14: 17588359221116605, 2022.
Article En | MEDLINE | ID: mdl-36032350

Introduction: Despite radical intent therapy for patients with stage III non-small-cell lung cancer (NSCLC), cumulative incidence of brain metastases (BM) reaches 30%. Current risk stratification methods fail to accurately identify these patients. As radiomics features have been shown to have predictive value, this study aims to develop a model combining clinical risk factors with radiomics features for BM development in patients with radically treated stage III NSCLC. Methods: Retrospective analysis of two prospective multicentre studies. Inclusion criteria: adequately staged [18F-fluorodeoxyglucose positron emission tomography-computed tomography (18-FDG-PET-CT), contrast-enhanced chest CT, contrast-enhanced brain magnetic resonance imaging/CT] and radically treated stage III NSCLC, exclusion criteria: second primary within 2 years of NSCLC diagnosis and prior prophylactic cranial irradiation. Primary endpoint was BM development any time during follow-up (FU). CT-based radiomics features (N = 530) were extracted from the primary lung tumour on 18-FDG-PET-CT images, and a list of clinical features (N = 8) was collected. Univariate feature selection based on the area under the curve (AUC) of the receiver operating characteristic was performed to identify relevant features. Generalized linear models were trained using the selected features, and multivariate predictive performance was assessed through the AUC. Results: In total, 219 patients were eligible for analysis. Median FU was 59.4 months for the training cohort and 67.3 months for the validation cohort; 21 (15%) and 17 (22%) patients developed BM in the training and validation cohort, respectively. Two relevant clinical features (age and adenocarcinoma histology) and four relevant radiomics features were identified as predictive. The clinical model yielded the highest AUC value of 0.71 (95% CI: 0.58-0.84), better than radiomics or a combination of clinical parameters and radiomics (both an AUC of 0.62, 95% CIs of 0.47-076 and 0.48-0.76, respectively). Conclusion: CT-based radiomics features of primary NSCLC in the current setup could not improve on a model based on clinical predictors (age and adenocarcinoma histology) of BM development in radically treated stage III NSCLC patients.

8.
Diagnostics (Basel) ; 12(7)2022 Jul 01.
Article En | MEDLINE | ID: mdl-35885513

Early diagnosis of COVID-19 is required to provide the best treatment to our patients, to prevent the epidemic from spreading in the community, and to reduce costs associated with the aggravation of the disease. We developed a decision tree model to evaluate the impact of using an artificial intelligence-based chest computed tomography (CT) analysis software (icolung, icometrix) to analyze CT scans for the detection and prognosis of COVID-19 cases. The model compared routine practice where patients receiving a chest CT scan were not screened for COVID-19, with a scenario where icolung was introduced to enable COVID-19 diagnosis. The primary outcome was to evaluate the impact of icolung on the transmission of COVID-19 infection, and the secondary outcome was the in-hospital length of stay. Using EUR 20000 as a willingness-to-pay threshold, icolung is cost-effective in reducing the risk of transmission, with a low prevalence of COVID-19 infections. Concerning the hospitalization cost, icolung is cost-effective at a higher value of COVID-19 prevalence and risk of hospitalization. This model provides a framework for the evaluation of AI-based tools for the early detection of COVID-19 cases. It allows for making decisions regarding their implementation in routine practice, considering both costs and effects.

9.
Radiology ; 305(2): 375-386, 2022 11.
Article En | MEDLINE | ID: mdl-35819326

Background Stratifying high-risk histopathologic features in endometrial carcinoma is important for treatment planning. Radiomics analysis at preoperative MRI holds potential to identify high-risk phenotypes. Purpose To evaluate the performance of multiparametric MRI three-dimensional radiomics-based machine learning models for differentiating low- from high-risk histopathologic markers-deep myometrial invasion (MI), lymphovascular space invasion (LVSI), and high-grade status-and advanced-stage endometrial carcinoma. Materials and Methods This dual-center retrospective study included women with histologically proven endometrial carcinoma who underwent 1.5-T MRI before hysterectomy between January 2011 and July 2015. Exclusion criteria were tumor diameter less than 1 cm, missing MRI sequences or histopathology reports, neoadjuvant therapy, and malignant neoplasms other than endometrial carcinoma. Three-dimensional radiomics features were extracted after tumor segmentation at MRI (T2-weighted, diffusion-weighted, and dynamic contrast-enhanced MRI). Predictive features were selected in the training set with use of random forest (RF) models for each end point, and trained RF models were applied to the external test set. Five board-certified radiologists conducted MRI-based staging and deep MI assessment in the training set. Areas under the receiver operating characteristic curve (AUCs) were reported with balanced accuracies, and radiologists' readings were compared with radiomics with use of McNemar tests. Results In total, 157 women were included: 94 at the first institution (training set; mean age, 66 years ± 11 [SD]) and 63 at the second institution (test set; 67 years ± 12). RF models dichotomizing deep MI, LVSI, high grade, and International Federation of Gynecology and Obstetrics (FIGO) stage led to AUCs of 0.81 (95% CI: 0.68, 0.88), 0.80 (95% CI: 0.67, 0.93), 0.74 (95% CI: 0.61, 0.86), and 0.84 (95% CI: 0.72, 0.92), respectively, in the test set. In the training set, radiomics provided increased performance compared with radiologists' readings for identifying deep MI (balanced accuracy, 86% vs 79%; P = .03), while no evidence of a difference was observed in performance for advanced FIGO stage (80% vs 78%; P = .27). Conclusion Three-dimensional radiomics can stratify patients by using preoperative MRI according to high-risk histopathologic end points in endometrial carcinoma and provide nonsignificantly different or higher performance than radiologists in identifying advanced stage and deep myometrial invasion, respectively. © RSNA, 2022 Online supplemental material is available for this article. See also the editorial by Kido and Nishio in this issue.


Endometrial Neoplasms , Multiparametric Magnetic Resonance Imaging , Humans , Female , Retrospective Studies , Endometrial Neoplasms/diagnostic imaging , Endometrial Neoplasms/surgery , Endometrial Neoplasms/pathology , Magnetic Resonance Imaging/methods , Risk Assessment
10.
Inf Fusion ; 82: 99-122, 2022 Jun.
Article En | MEDLINE | ID: mdl-35664012

Removing the bias and variance of multicentre data has always been a challenge in large scale digital healthcare studies, which requires the ability to integrate clinical features extracted from data acquired by different scanners and protocols to improve stability and robustness. Previous studies have described various computational approaches to fuse single modality multicentre datasets. However, these surveys rarely focused on evaluation metrics and lacked a checklist for computational data harmonisation studies. In this systematic review, we summarise the computational data harmonisation approaches for multi-modality data in the digital healthcare field, including harmonisation strategies and evaluation metrics based on different theories. In addition, a comprehensive checklist that summarises common practices for data harmonisation studies is proposed to guide researchers to report their research findings more effectively. Last but not least, flowcharts presenting possible ways for methodology and metric selection are proposed and the limitations of different methods have been surveyed for future research.

11.
Comput Biol Med ; 140: 105111, 2021 Dec 04.
Article En | MEDLINE | ID: mdl-34891095

Artificial Intelligence (AI) has emerged as a useful aid in numerous clinical applications for diagnosis and treatment decisions. Deep neural networks have shown the same or better performance than clinicians in many tasks owing to the rapid increase in the available data and computational power. In order to conform to the principles of trustworthy AI, it is essential that the AI system be transparent, robust, fair, and ensure accountability. Current deep neural solutions are referred to as black-boxes due to a lack of understanding of the specifics concerning the decision-making process. Therefore, there is a need to ensure the interpretability of deep neural networks before they can be incorporated into the routine clinical workflow. In this narrative review, we utilized systematic keyword searches and domain expertise to identify nine different types of interpretability methods that have been used for understanding deep learning models for medical image analysis applications based on the type of generated explanations and technical similarities. Furthermore, we report the progress made towards evaluating the explanations produced by various interpretability methods. Finally, we discuss limitations, provide guidelines for using interpretability methods and future directions concerning the interpretability of deep neural networks for medical imaging analysis.

12.
J Pers Med ; 11(9)2021 Aug 27.
Article En | MEDLINE | ID: mdl-34575619

Radiomics converts medical images into mineable data via a high-throughput extraction of quantitative features used for clinical decision support. However, these radiomic features are susceptible to variation across scanners, acquisition protocols, and reconstruction settings. Various investigations have assessed the reproducibility and validation of radiomic features across these discrepancies. In this narrative review, we combine systematic keyword searches with prior domain knowledge to discuss various harmonization solutions to make the radiomic features more reproducible across various scanners and protocol settings. Different harmonization solutions are discussed and divided into two main categories: image domain and feature domain. The image domain category comprises methods such as the standardization of image acquisition, post-processing of raw sensor-level image data, data augmentation techniques, and style transfer. The feature domain category consists of methods such as the identification of reproducible features and normalization techniques such as statistical normalization, intensity harmonization, ComBat and its derivatives, and normalization using deep learning. We also reflect upon the importance of deep learning solutions for addressing variability across multi-centric radiomic studies especially using generative adversarial networks (GANs), neural style transfer (NST) techniques, or a combination of both. We cover a broader range of methods especially GANs and NST methods in more detail than previous reviews.

13.
Cancers (Basel) ; 13(15)2021 Jul 24.
Article En | MEDLINE | ID: mdl-34359623

Current radiomic studies of head and neck squamous cell carcinomas (HNSCC) are typically based on datasets combining tumors from different locations, assuming that the radiomic features are similar based on histopathologic characteristics. However, molecular pathogenesis and treatment in HNSCC substantially vary across different tumor sites. It is not known if a statistical difference exists between radiomic features from different tumor sites and how they affect machine learning model performance in endpoint prediction. To answer these questions, we extracted radiomic features from contrast-enhanced neck computed tomography scans (CTs) of 605 patients with HNSCC originating from the oral cavity, oropharynx, and hypopharynx/larynx. The difference in radiomic features of tumors from these sites was assessed using statistical analyses and Random Forest classifiers on the radiomic features with 10-fold cross-validation to predict tumor sites, nodal metastasis, and HPV status. We found statistically significant differences (p-value ≤ 0.05) between the radiomic features of HNSCC depending on tumor location. We also observed that differences in quantitative features among HNSCC from different locations impact the performance of machine learning models. This suggests that radiomic features may reveal biologic heterogeneity complementary to current gold standard histopathologic evaluation. We recommend considering tumor site in radiomic studies of HNSCC.

14.
Front Neurosci ; 15: 679941, 2021.
Article En | MEDLINE | ID: mdl-34421515

Conventional magnetic resonance imaging (cMRI) is poorly sensitive to pathological changes related to multiple sclerosis (MS) in normal-appearing white matter (NAWM) and gray matter (GM), with the added difficulty of not being very reproducible. Quantitative MRI (qMRI), on the other hand, attempts to represent the physical properties of tissues, making it an ideal candidate for quantitative medical image analysis or radiomics. We therefore hypothesized that qMRI-based radiomic features have added diagnostic value in MS compared to cMRI. This study investigated the ability of cMRI (T1w) and qMRI features extracted from white matter (WM), NAWM, and GM to distinguish between MS patients (MSP) and healthy control subjects (HCS). We developed exploratory radiomic classification models on a dataset comprising 36 MSP and 36 HCS recruited in CHU Liege, Belgium, acquired with cMRI and qMRI. For each image type and region of interest, qMRI radiomic models for MS diagnosis were developed on a training subset and validated on a testing subset. Radiomic models based on cMRI were developed on the entire training dataset and externally validated on open-source datasets with 167 HCS and 10 MSP. Ranked by region of interest, the best diagnostic performance was achieved in the whole WM. Here the model based on magnetization transfer imaging (a type of qMRI) features yielded a median area under the receiver operating characteristic curve (AUC) of 1.00 in the testing sub-cohort. Ranked by image type, the best performance was achieved by the magnetization transfer models, with median AUCs of 0.79 (0.69-0.90, 90% CI) in NAWM and 0.81 (0.71-0.90) in GM. The external validation of the T1w models yielded an AUC of 0.78 (0.47-1.00) in the whole WM, demonstrating a large 95% CI and a low sensitivity of 0.30 (0.10-0.70). This exploratory study indicates that qMRI radiomics could provide efficient diagnostic information using NAWM and GM analysis in MSP. T1w radiomics could be useful for a fast and automated check of conventional MRI for WM abnormalities once acquisition and reconstruction heterogeneities have been overcome. Further prospective validation is needed, involving more data for better interpretation and generalization of the results.

15.
BJR Open ; 2(1): 20210006, 2021.
Article En | MEDLINE | ID: mdl-34381940

OBJECTIVES: Compare a quantitative, algorithm-driven, and qualitative, pathologist-driven, scoring of radiation-induced pulmonary fibrosis (RIPF). And using these scoring models to derive preliminary comparisons on the effects of different mesenchymal stem cell (MSC) administration modalities in reducing RIPF. METHODS: 25 rats were randomized into 5 groups: non-irradiated control (CG), irradiated control (CR), intraperitoneally administered granulocyte-macrophage colony stimulating factor or GM-CSF (Drug), intravascularly administered MSC (IV), and intratracheally administered MSC (IT). All groups, except CG, received an 18 Gy conformal dose to the right lung. Drug, IV and IT groups were treated immediately after irradiation. After 24 weeks of observation, rats were euthanized, their lungs excised, fixed and stained with Masson's Trichrome. Samples were anonymized and RIPF was scored qualitatively by a certified pathologist and quantitatively using ImageScope. An analysis of association was conducted, and two binary classifiers trained to validate the integrity of both qualitative and quantitative scoring. Differences between the treatment groups, as assessed by the pathologist score, were then tested by variance component analysis and mixed models for differences in RIPF outcomes. RESULTS: There is agreement between qualitative and quantitative scoring for RIPF grades from 4 to 7. Both classifiers performed similarly on the testing set (AUC = 0.923) indicating accordance between the qualitative and quantitative scoring. For comparisons between MSC infusion modalities, the Drug group had better outcomes (mean pathologist scoring of 3.96), correlating with significantly better RIPF outcomes than IV [lower by 0.97, p = 0.047, 95% CI = (0.013, 1.918)] and resulting in an improvement over CR [lower by 0.93, p = 0.037, 95% CI = (0.062, 1.800]. CONCLUSION: Quantitative image analysis may help in the assessment of therapeutic interventions for RIPF and can serve as a scoring surrogate in differentiating between severe and mild cases of RIPF. Preliminary data demonstrate that the use of GM-CSF was best correlated with lower RIPF severity. ADVANCES IN KNOWLEDGE: Quantitative image analysis can be a viable supplemental system of quality control and triaging in situations where pathologist work hours or resources are limited. The use of different MSC administration methods can result in different degrees of MSC efficacy and study outcomes.

16.
Phys Med ; 88: 272-277, 2021 Aug.
Article En | MEDLINE | ID: mdl-34329921

PURPOSE: Radiomic texture calculation requires discretizing image intensities within the region-of-interest. FBN (fixed-bin-number), FBS (fixed-bin-size) and FBN and FBS with intensity equalization (FBNequal, FBSequal) are four discretization approaches. A crucial choice is the voxel intensity (Hounsfield units, or HU) binning range. We assessed the effect of this choice on radiomic features. METHODS: The dataset comprised 95 patients with head-and-neck squamous-cell-carcinoma. Dual energy CT data was reconstructed at 21 electron energies (40, 45,… 140 keV). Each of 94 texture features were calculated with 64 extraction parameters. All features were calculated five times: original choice, left shift (-10/-20 HU), right shift (+10/+20 HU). For each feature, Spearman correlation between nominal and four variants were calculated to determine feature stability. This was done for six texture feature types (GLCM, GLRLM, GLSZM, GLDZM, NGTDM, and NGLDM) separately. This analysis was repeated for the four binning algorithms. Effect of feature instability on predictive ability was studied for lymphadenopathy as endpoint. RESULTS: FBN and FBNequal algorithms showed good stability (correlation values consistently >0.9). For FBS and FBSequal algorithms, while median values exceeded 0.9, the 95% lower bound decreased as a function of energy, with poor performance over the entire spectrum. FBNequal was the most stable algorithm, and FBS the least. CONCLUSIONS: We believe this is the first multi-energy systematic study of the impact of CT HU range used during intensity discretization for radiomic feature extraction. Future analyses should account for this source of uncertainty when evaluating the robustness of their radiomic signature.


Algorithms , Image Processing, Computer-Assisted , Humans , Tomography, X-Ray Computed
17.
Cancers (Basel) ; 13(10)2021 May 18.
Article En | MEDLINE | ID: mdl-34070016

This retrospective study investigated the value of pretreatment contrast-enhanced Magnetic Resonance Imaging (MRI)-based radiomics for the prediction of pathologic complete tumor response to neoadjuvant systemic therapy in breast cancer patients. A total of 292 breast cancer patients, with 320 tumors, who were treated with neo-adjuvant systemic therapy and underwent a pretreatment MRI exam were enrolled. As the data were collected in two different hospitals with five different MRI scanners and varying acquisition protocols, three different strategies to split training and validation datasets were used. Radiomics, clinical, and combined models were developed using random forest classifiers in each strategy. The analysis of radiomics features had no added value in predicting pathologic complete tumor response to neoadjuvant systemic therapy in breast cancer patients compared with the clinical models, nor did the combined models perform significantly better than the clinical models. Further, the radiomics features selected for the models and their performance differed with and within the different strategies. Due to previous and current work, we tentatively attribute the lack of improvement in clinical models following the addition of radiomics to the effects of variations in acquisition and reconstruction parameters. The lack of reproducibility data (i.e., test-retest or similar) meant that this effect could not be analyzed. These results indicate the need for reproducibility studies to preselect reproducible features in order to properly assess the potential of radiomics.

18.
Cancers (Basel) ; 13(11)2021 May 29.
Article En | MEDLINE | ID: mdl-34072509

The aim of this study is to build a decision support system (DSS) to select radical prostatectomy (RP) or external beam radiotherapy (EBRT) for low- to intermediate-risk prostate cancer patients. We used an individual state-transition model based on predictive models for estimating tumor control and toxicity probabilities. We performed analyses on a synthetically generated dataset of 1000 patients with realistic clinical parameters, externally validated by comparison to randomized clinical trials, and set up an in silico clinical trial for elderly patients. We assessed the cost-effectiveness (CE) of the DSS for treatment selection by comparing it to randomized treatment allotment. Using the DSS, 47.8% of synthetic patients were selected for RP and 52.2% for EBRT. During validation, differences with the simulations of late toxicity and biochemical failure never exceeded 2%. The in silico trial showed that for elderly patients, toxicity has more influence on the decision than TCP, and the predicted QoL depends on the initial erectile function. The DSS is estimated to result in cost savings (EUR 323 (95% CI: EUR 213-433)) and more quality-adjusted life years (QALYs; 0.11 years, 95% CI: 0.00-0.22) than randomized treatment selection.

20.
PLoS One ; 16(4): e0249920, 2021.
Article En | MEDLINE | ID: mdl-33857224

OBJECTIVE: To establish whether one can build a mortality prediction model for COVID-19 patients based solely on demographics and comorbidity data that outperforms age alone. Such a model could be a precursor to implementing smart lockdowns and vaccine distribution strategies. METHODS: The training cohort comprised 2337 COVID-19 inpatients from nine hospitals in The Netherlands. The clinical outcome was death within 21 days of being discharged. The features were derived from electronic health records collected during admission. Three feature selection methods were used: LASSO, univariate using a novel metric, and pairwise (age being half of each pair). 478 patients from Belgium were used to test the model. All modeling attempts were compared against an age-only model. RESULTS: In the training cohort, the mortality group's median age was 77 years (interquartile range = 70-83), higher than the non-mortality group (median = 65, IQR = 55-75). The incidence of former/active smokers, male gender, hypertension, diabetes, dementia, cancer, chronic obstructive pulmonary disease, chronic cardiac disease, chronic neurological disease, and chronic kidney disease was higher in the mortality group. All stated differences were statistically significant after Bonferroni correction. LASSO selected eight features, novel univariate chose five, and pairwise chose none. No model was able to surpass an age-only model in the external validation set, where age had an AUC of 0.85 and a balanced accuracy of 0.77. CONCLUSION: When applied to an external validation set, we found that an age-only mortality model outperformed all modeling attempts (curated on www.covid19risk.ai) using three feature selection methods on 22 demographic and comorbid features.


COVID-19/mortality , Age Factors , Aged , Aged, 80 and over , Belgium/epidemiology , COVID-19/diagnosis , COVID-19/epidemiology , Cohort Studies , Communicable Disease Control , Comorbidity , Electronic Health Records , Female , Hospitalization , Humans , Male , Middle Aged , Netherlands/epidemiology , Prognosis , Risk Assessment , Risk Factors , SARS-CoV-2/isolation & purification
...