Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 509
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Oncologist ; 29(2): e187-e197, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-37669223

ABSTRACT

BACKGROUND: Not only should resistance to neoadjuvant chemotherapy (NAC) be considered in patients with breast cancer but also the possibility of achieving a pathologic complete response (PCR) after NAC. Our study aims to develop 2 multimodal ultrasound deep learning (DL) models to noninvasively predict resistance and PCR to NAC before treatment. METHODS: From January 2017 to July 2022, a total of 170 patients with breast cancer were prospectively enrolled. All patients underwent multimodal ultrasound examination (grayscale 2D ultrasound and ultrasound elastography) before NAC. We combined clinicopathological information to develop 2 DL models, DL_Clinical_resistance and DL_Clinical_PCR, for predicting resistance and PCR to NAC, respectively. In addition, these 2 models were combined to stratify the prediction of response to NAC. RESULTS: In the test cohort, DL_Clinical_resistance had an AUC of 0.911 (95%CI, 0.814-0.979) with a sensitivity of 0.905 (95%CI, 0.765-1.000) and an NPV of 0.882 (95%CI, 0.708-1.000). Meanwhile, DL_Clinical_PCR achieved an AUC of 0.880 (95%CI, 0.751-0.973) and sensitivity and NPV of 0.875 (95%CI, 0.688-1.000) and 0.895 (95%CI, 0.739-1.000), respectively. By combining DL_Clinical_resistance and DL_Clinical_PCR, 37.1% of patients with resistance and 25.7% of patients with PCR were successfully identified by the combined model, suggesting that these patients could benefit by an early change of treatment strategy or by implementing an organ preservation strategy after NAC. CONCLUSIONS: The proposed DL_Clinical_resistance and DL_Clinical_PCR models and combined strategy have the potential to predict resistance and PCR to NAC before treatment and allow stratified prediction of NAC response.


Subject(s)
Breast Neoplasms , Deep Learning , Humans , Female , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Neoadjuvant Therapy , Retrospective Studies
2.
Crit Rev Microbiol ; : 1-16, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38346140

ABSTRACT

Cancer immunotherapies have been widely hailed as a breakthrough for cancer treatment in the last decade, epitomized by the unprecedented results observed with checkpoint blockade. Even so, only a minority of patients currently achieve durable remissions. In general, responsive patients appear to have either a high number of tumor neoantigens, a preexisting immune cell infiltrate in the tumor microenvironment, or an 'immune-active' transcriptional profile, determined in part by the presence of a type I interferon gene signature. These observations suggest that the therapeutic efficacy of immunotherapy can be enhanced through strategies that release tumor neoantigens and/or produce a pro-inflammatory tumor microenvironment. In principle, exogenous tumor-targeting bacteria offer a unique solution for improving responsiveness to immunotherapy. This review discusses how tumor-selective bacterial infection can modulate the immunological microenvironment of the tumor and the potential for combination with cancer immunotherapy strategies to further increase therapeutic efficacy. In addition, we provide a perspective on the clinical translation of replicating bacterial therapies, with a focus on the challenges that must be resolved to ensure a successful outcome.

3.
Microb Cell Fact ; 23(1): 119, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38659027

ABSTRACT

BACKGROUND: Clostridium spp. has demonstrated therapeutic potential in cancer treatment through intravenous or intratumoral administration. This approach has expanded to include non-pathogenic clostridia for the treatment of various diseases, underscoring the innovative concept of oral-spore vaccination using clostridia. Recent advancements in the field of synthetic biology have significantly enhanced the development of Clostridium-based bio-therapeutics. These advancements are particularly notable in the areas of efficient protein overexpression and secretion, which are crucial for the feasibility of oral vaccination strategies. Here, we present two examples of genetically engineered Clostridium candidates: one as an oral cancer vaccine and the other as an antiviral oral vaccine against SARS-CoV-2. RESULTS: Using five validated promoters and a signal peptide derived from Clostridium sporogenes, a series of full-length NY-ESO-1/CTAG1, a promising cancer vaccine candidate, expression vectors were constructed and transformed into C. sporogenes and Clostridium butyricum. Western blotting analysis confirmed efficient expression and secretion of NY-ESO-1 in clostridia, with specific promoters leading to enhanced detection signals. Additionally, the fusion of a reported bacterial adjuvant to NY-ESO-1 for improved immune recognition led to the cloning difficulties in E. coli. The use of an AUU start codon successfully mitigated potential toxicity issues in E. coli, enabling the secretion of recombinant proteins in C. sporogenes and C. butyricum. We further demonstrate the successful replacement of PyrE loci with high-expression cassettes carrying NY-ESO-1 and adjuvant-fused NY-ESO-1, achieving plasmid-free clostridia capable of secreting the antigens. Lastly, the study successfully extends its multiplex genetic manipulations to engineer clostridia for the secretion of SARS-CoV-2-related Spike_S1 antigens. CONCLUSIONS: This study successfully demonstrated that C. butyricum and C. sporogenes can produce the two recombinant antigen proteins (NY-ESO-1 and SARS-CoV-2-related Spike_S1 antigens) through genetic manipulations, utilizing the AUU start codon. This approach overcomes challenges in cloning difficult proteins in E. coli. These findings underscore the feasibility of harnessing commensal clostridia for antigen protein secretion, emphasizing the applicability of non-canonical translation initiation across diverse species with broad implications for medical or industrial biotechnology.


Subject(s)
Clostridium butyricum , Clostridium , Recombinant Proteins , Clostridium butyricum/genetics , Clostridium butyricum/metabolism , Clostridium/genetics , Clostridium/metabolism , Humans , Recombinant Proteins/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/genetics , Cancer Vaccines/immunology , Cancer Vaccines/genetics , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Administration, Oral , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Spores, Bacterial/genetics , Spores, Bacterial/immunology , Vaccination , COVID-19/prevention & control , Genetic Engineering , Escherichia coli/genetics , Escherichia coli/metabolism , Promoter Regions, Genetic
4.
Radiology ; 307(5): e221843, 2023 06.
Article in English | MEDLINE | ID: mdl-37338353

ABSTRACT

Background Handcrafted radiomics and deep learning (DL) models individually achieve good performance in lesion classification (benign vs malignant) on contrast-enhanced mammography (CEM) images. Purpose To develop a comprehensive machine learning tool able to fully automatically identify, segment, and classify breast lesions on the basis of CEM images in recall patients. Materials and Methods CEM images and clinical data were retrospectively collected between 2013 and 2018 for 1601 recall patients at Maastricht UMC+ and 283 patients at Gustave Roussy Institute for external validation. Lesions with a known status (malignant or benign) were delineated by a research assistant overseen by an expert breast radiologist. Preprocessed low-energy and recombined images were used to train a DL model for automatic lesion identification, segmentation, and classification. A handcrafted radiomics model was also trained to classify both human- and DL-segmented lesions. Sensitivity for identification and the area under the receiver operating characteristic curve (AUC) for classification were compared between individual and combined models at the image and patient levels. Results After the exclusion of patients without suspicious lesions, the total number of patients included in the training, test, and validation data sets were 850 (mean age, 63 years ± 8 [SD]), 212 (62 years ± 8), and 279 (55 years ± 12), respectively. In the external data set, lesion identification sensitivity was 90% and 99% at the image and patient level, respectively, and the mean Dice coefficient was 0.71 and 0.80 at the image and patient level, respectively. Using manual segmentations, the combined DL and handcrafted radiomics classification model achieved the highest AUC (0.88 [95% CI: 0.86, 0.91]) (P < .05 except compared with DL, handcrafted radiomics, and clinical features model, where P = .90). Using DL-generated segmentations, the combined DL and handcrafted radiomics model showed the highest AUC (0.95 [95% CI: 0.94, 0.96]) (P < .05). Conclusion The DL model accurately identified and delineated suspicious lesions on CEM images, and the combined output of the DL and handcrafted radiomics models achieved good diagnostic performance. © RSNA, 2023 Supplemental material is available for this article. See also the editorial by Bahl and Do in this issue.


Subject(s)
Deep Learning , Humans , Middle Aged , Retrospective Studies , Mammography/methods , Breast/diagnostic imaging , ROC Curve
5.
Med Res Rev ; 42(1): 426-440, 2022 01.
Article in English | MEDLINE | ID: mdl-34309893

ABSTRACT

Radiomics is the quantitative analysis of standard-of-care medical imaging; the information obtained can be applied within clinical decision support systems to create diagnostic, prognostic, and/or predictive models. Radiomics analysis can be performed by extracting hand-crafted radiomics features or via deep learning algorithms. Radiomics has evolved tremendously in the last decade, becoming a bridge between imaging and precision medicine. Radiomics exploits sophisticated image analysis tools coupled with statistical elaboration to extract the wealth of information hidden inside medical images, such as computed tomography (CT), magnetic resonance (MR), and/or Positron emission tomography (PET) scans, routinely performed in the everyday clinical practice. Many efforts have been devoted in recent years to the standardization and validation of radiomics approaches, to demonstrate their usefulness and robustness beyond any reasonable doubts. However, the booming of publications and commercial applications of radiomics approaches warrant caution and proper understanding of all the factors involved to avoid "scientific pollution" and overly enthusiastic claims by researchers and clinicians alike. For these reasons the present review aims to be a guidebook of sorts, describing the process of radiomics, its pitfalls, challenges, and opportunities, along with its ability to improve clinical decision-making, from oncology and respiratory medicine to pharmacological and genotyping studies.


Subject(s)
Image Processing, Computer-Assisted , Precision Medicine , Humans , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging , Medical Oncology , Positron-Emission Tomography
6.
Eur J Nucl Med Mol Imaging ; 49(13): 4452-4463, 2022 11.
Article in English | MEDLINE | ID: mdl-35809090

ABSTRACT

Artificial intelligence (AI) will change the face of nuclear medicine and molecular imaging as it will in everyday life. In this review, we focus on the potential applications of AI in the field, both from a physical (radiomics, underlying statistics, image reconstruction and data analysis) and a clinical (neurology, cardiology, oncology) perspective. Challenges for transferability from research to clinical practice are being discussed as is the concept of explainable AI. Finally, we focus on the fields where challenges should be set out to introduce AI in the field of nuclear medicine and molecular imaging in a reliable manner.


Subject(s)
Artificial Intelligence , Nuclear Medicine , Humans , Radionuclide Imaging , Image Processing, Computer-Assisted/methods , Molecular Imaging
7.
Eur Radiol ; 32(4): 2188-2199, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34842959

ABSTRACT

OBJECTIVES: An accurate and rapid diagnosis is crucial for the appropriate treatment of pulmonary tuberculosis (TB). This study aims to develop an artificial intelligence (AI)-based fully automated CT image analysis system for detection, diagnosis, and burden quantification of pulmonary TB. METHODS: From December 2007 to September 2020, 892 chest CT scans from pathogen-confirmed TB patients were retrospectively included. A deep learning-based cascading framework was connected to create a processing pipeline. For training and validation of the model, 1921 lesions were manually labeled, classified according to six categories of critical imaging features, and visually scored regarding lesion involvement as the ground truth. A "TB score" was calculated based on a network-activation map to quantitively assess the disease burden. Independent testing datasets from two additional hospitals (dataset 2, n = 99; dataset 3, n = 86) and the NIH TB Portals (n = 171) were used to externally validate the performance of the AI model. RESULTS: CT scans of 526 participants (mean age, 48.5 ± 16.5 years; 206 women) were analyzed. The lung lesion detection subsystem yielded a mean average precision of the validation cohort of 0.68. The overall classification accuracy of six pulmonary critical imaging findings indicative of TB of the independent datasets was 81.08-91.05%. A moderate to strong correlation was demonstrated between the AI model-quantified TB score and the radiologist-estimated CT score. CONCLUSIONS: The proposed end-to-end AI system based on chest CT can achieve human-level diagnostic performance for early detection and optimal clinical management of patients with pulmonary TB. KEY POINTS: • Deep learning allows automatic detection, diagnosis, and evaluation of pulmonary tuberculosis. • Artificial intelligence helps clinicians to assess patients with tuberculosis. • Pulmonary tuberculosis disease activity and treatment management can be improved.


Subject(s)
Artificial Intelligence , Tuberculosis, Pulmonary , Adult , Aged , Female , Humans , Image Processing, Computer-Assisted , Middle Aged , Retrospective Studies , Tomography, X-Ray Computed/methods , Tuberculosis, Pulmonary/diagnostic imaging
8.
Urol Int ; 106(1): 63-74, 2022.
Article in English | MEDLINE | ID: mdl-34130300

ABSTRACT

OBJECTIVE: The purpose of this review was to summarize the current literature on the assessment and treatment of radiation urethritis and cystitis (RUC) for the development of an evidenced-based management algorithm. MATERIAL AND METHODS: The PubMed/MEDLINE database was searched by a multidisciplinary group of experts in January 2021. RESULTS: In total, 48 publications were identified. Three different types of RUC can be observed in clinical practice: inflammation-predominant, bleeding-predominant, and the combination of inflammation- and bleeding-RUC. There is no consensus on the optimal treatment of RUC. Inflammation-predominant RUC should be treated symptomatically based on the existence of bothersome storage or voiding lower urinary tract symptom as well as on pain. When bleeding-predominant RUC has occurred, hydration and hyperbaric oxygen therapy (HOT) should be used first and, if HOT is not available, oral drugs instead (sodium pentosane polysulfate, aminocaproic acid, immunokine WF 10, conjugated estrogene, or pentoxifylline + vitamin E). If local bleeding persists, focal therapy of bleeding vessels with a laser or electrocoagulation is indicated. In case of generalized bleeding, intravesical installation should be initiated (formalin, aluminium salts, and hyaluronic acid/chondroitin). Vessel embolization is a less invasive treatment with potentially less complications and good clinical outcomes. Open- or robot-assisted surgery is indicated in patients with permanent, life-threatening bleeding, or fistulae. CONCLUSIONS: Treatment of RUC, if not self-limiting, should be done according to the type of RUC and in a stepwise approach. Conservative/medical treatment (oral and topic agents) should primarily be used before invasive (transurethral) treatments.


Subject(s)
Algorithms , Cystitis/diagnosis , Cystitis/therapy , Radiation Injuries/diagnosis , Radiation Injuries/therapy , Urethritis/diagnosis , Urethritis/therapy , Acute Disease , Chronic Disease , Humans
9.
Radiol Med ; 127(1): 72-82, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34822101

ABSTRACT

PURPOSE: This study investigated the feasibility of a new image analysis technique (radiomics) on conventional MRI for the computer-aided diagnosis of Menière's disease. MATERIALS AND METHODS: A retrospective, multicentric diagnostic case-control study was performed. This study included 120 patients with unilateral or bilateral Menière's disease and 140 controls from four centers in the Netherlands and Belgium. Multiple radiomic features were extracted from conventional MRI scans and used to train a machine learning-based, multi-layer perceptron classification model to distinguish patients with Menière's disease from controls. The primary outcomes were accuracy, sensitivity, specificity, positive predictive value, and negative predictive value of the classification model. RESULTS: The classification accuracy of the machine learning model on the test set was 82%, with a sensitivity of 83%, and a specificity of 82%. The positive and negative predictive values were 71%, and 90%, respectively. CONCLUSION: The multi-layer perceptron classification model yielded a precise, high-diagnostic performance in identifying patients with Menière's disease based on radiomic features extracted from conventional T2-weighted MRI scans. In the future, radiomics might serve as a fast and noninvasive decision support system, next to clinical evaluation in the diagnosis of Menière's disease.


Subject(s)
Image Interpretation, Computer-Assisted/methods , Machine Learning , Magnetic Resonance Imaging/methods , Meniere Disease/diagnostic imaging , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Ear, Inner/diagnostic imaging , Feasibility Studies , Female , Humans , Male , Middle Aged , Retrospective Studies , Young Adult
10.
Inf Fusion ; 82: 99-122, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35664012

ABSTRACT

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.
Radiology ; 298(2): 248-260, 2021 02.
Article in English | MEDLINE | ID: mdl-33350894

ABSTRACT

Radiation therapy (RT) continues to be one of the mainstays of cancer treatment. Considerable efforts have been recently devoted to integrating MRI into clinical RT planning and monitoring. This integration, known as MRI-guided RT, has been motivated by the superior soft-tissue contrast, organ motion visualization, and ability to monitor tumor and tissue physiologic changes provided by MRI compared with CT. Offline MRI is already used for treatment planning at many institutions. Furthermore, MRI-guided linear accelerator systems, allowing use of MRI during treatment, enable improved adaptation to anatomic changes between RT fractions compared with CT guidance. Efforts are underway to develop real-time MRI-guided intrafraction adaptive RT of tumors affected by motion and MRI-derived biomarkers to monitor treatment response and potentially adapt treatment to physiologic changes. These developments in MRI guidance provide the basis for a paradigm change in treatment planning, monitoring, and adaptation. Key challenges to advancing MRI-guided RT include real-time volumetric anatomic imaging, addressing image distortion because of magnetic field inhomogeneities, reproducible quantitative imaging across different MRI systems, and biologic validation of quantitative imaging. This review describes emerging innovations in offline and online MRI-guided RT, exciting opportunities they offer for advancing research and clinical care, hurdles to be overcome, and the need for multidisciplinary collaboration.


Subject(s)
Magnetic Resonance Imaging/methods , Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Radiation Oncology/methods , Radiology, Interventional/methods , Humans
12.
Eur J Nucl Med Mol Imaging ; 48(12): 3961-3974, 2021 11.
Article in English | MEDLINE | ID: mdl-33693966

ABSTRACT

INTRODUCTION: Lung cancer ranks second in new cancer cases and first in cancer-related deaths worldwide. Precision medicine is working on altering treatment approaches and improving outcomes in this patient population. Radiological images are a powerful non-invasive tool in the screening and diagnosis of early-stage lung cancer, treatment strategy support, prognosis assessment, and follow-up for advanced-stage lung cancer. Recently, radiological features have evolved from solely semantic to include (handcrafted and deep) radiomic features. Radiomics entails the extraction and analysis of quantitative features from medical images using mathematical and machine learning methods to explore possible ties with biology and clinical outcomes. METHODS: Here, we outline the latest applications of both structural and functional radiomics in detection, diagnosis, and prediction of pathology, gene mutation, treatment strategy, follow-up, treatment response evaluation, and prognosis in the field of lung cancer. CONCLUSION: The major drawbacks of radiomics are the lack of large datasets with high-quality data, standardization of methodology, the black-box nature of deep learning, and reproducibility. The prerequisite for the clinical implementation of radiomics is that these limitations are addressed. Future directions include a safer and more efficient model-training mode, merge multi-modality images, and combined multi-discipline or multi-omics to form "Medomics."


Subject(s)
Lung Neoplasms , Humans , Lung , Lung Neoplasms/diagnostic imaging , Machine Learning , Prognosis , Reproducibility of Results
13.
Eur J Nucl Med Mol Imaging ; 48(11): 3432-3443, 2021 10.
Article in English | MEDLINE | ID: mdl-33772334

ABSTRACT

PURPOSE: To test the performances of native and tumour to liver ratio (TLR) radiomic features extracted from pre-treatment 2-[18F] fluoro-2-deoxy-D-glucose ([18F]FDG) PET/CT and combined with machine learning (ML) for predicting cancer recurrence in patients with locally advanced cervical cancer (LACC). METHODS: One hundred fifty-eight patients with LACC from multiple centers were retrospectively included in the study. Tumours were segmented using the Fuzzy Local Adaptive Bayesian (FLAB) algorithm. Radiomic features were extracted from the tumours and from regions drawn over the normal liver. Cox proportional hazard model was used to test statistical significance of clinical and radiomic features. Fivefold cross validation was used to tune the number of features. Seven different feature selection methods and four classifiers were tested. The models with the selected features were trained using bootstrapping and tested in data from each scanner independently. Reproducibility of radiomics features, clinical data added value and effect of ComBat-based harmonisation were evaluated across scanners. RESULTS: After a median follow-up of 23 months, 29% of the patients recurred. No individual radiomic or clinical features were significantly associated with cancer recurrence. The best model was obtained using 10 TLR features combined with clinical information. The area under the curve (AUC), F1-score, precision and recall were respectively 0.78 (0.67-0.88), 0.49 (0.25-0.67), 0.42 (0.25-0.60) and 0.63 (0.20-0.80). ComBat did not improve the predictive performance of the best models. Both the TLR and the native models performance varied across scanners used in the test set. CONCLUSION: [18F]FDG PET radiomic features combined with ML add relevant information to the standard clinical parameters in terms of LACC patient's outcome but remain subject to variability across PET/CT devices.


Subject(s)
Fluorodeoxyglucose F18 , Uterine Cervical Neoplasms , Bayes Theorem , Disease-Free Survival , Female , Humans , Neoplasm Recurrence, Local , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals , Reproducibility of Results , Retrospective Studies , Uterine Cervical Neoplasms/diagnostic imaging
14.
PLoS Comput Biol ; 16(8): e1008041, 2020 08.
Article in English | MEDLINE | ID: mdl-32745136

ABSTRACT

Hypoxia-activated prodrugs (HAPs) present a conceptually elegant approach to not only overcome, but better yet, exploit intra-tumoural hypoxia. Despite being successful in vitro and in vivo, HAPs are yet to achieve successful results in clinical settings. It has been hypothesised that this lack of clinical success can, in part, be explained by the insufficiently stringent clinical screening selection of determining which tumours are suitable for HAP treatments. Taking a mathematical modelling approach, we investigate how tumour properties and HAP-radiation scheduling influence treatment outcomes in simulated tumours. The following key results are demonstrated in silico: (i) HAP and ionising radiation (IR) monotherapies may attack tumours in dissimilar, and complementary, ways. (ii) HAP-IR scheduling may impact treatment efficacy. (iii) HAPs may function as IR treatment intensifiers. (iv) The spatio-temporal intra-tumoural oxygen landscape may impact HAP efficacy. Our in silico framework is based on an on-lattice, hybrid, multiscale cellular automaton spanning three spatial dimensions. The mathematical model for tumour spheroid growth is parameterised by multicellular tumour spheroid (MCTS) data.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Hypoxia/physiology , Models, Biological , Prodrugs/pharmacology , Tumor Microenvironment/physiology , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Computational Biology , Computer Simulation , Humans , Radiation, Ionizing , Radiotherapy , Spheroids, Cellular , Tumor Cells, Cultured
15.
Esophagus ; 18(1): 100-110, 2021 01.
Article in English | MEDLINE | ID: mdl-32889674

ABSTRACT

BACKGROUND: The presence of lymph node metastasis (LNmets) is a poor prognostic factor in oesophageal cancer (OeC) patients treated with neoadjuvant chemoradiotherapy (nCRT) followed by surgery. Tumour regression grade (TRG) in LNmets has been suggested as a predictor for survival. The aim of this study was to investigate whether TRG in LNmets is related to their location within the radiotherapy (RT) field. METHODS: Histopathological TRG was retrospectively classified in 2565 lymph nodes (LNs) from 117 OeC patients treated with nCRT and surgery as: (A) no tumour, no signs of regression; (B) tumour without regression; (C) viable tumour and regression; and (D) complete response. Multivariate survival analysis was used to investigate the relationship between LN location within the RT field, pathological TRG of the LN and TRG of the primary tumour. RESULTS: In 63 (54%) patients, viable tumour cells or signs of regression were seen in 264 (10.2%) LNs which were classified as TRG-B (n = 56), C (n = 104) or D (n = 104) LNs. 73% of B, C and D LNs were located within the RT field. There was a trend towards a relationship between LN response and anatomical LN location with respect to the RT field (p = 0.052). Multivariate analysis showed that only the presence of LNmets within the RT field with TRG-B is related to poor overall survival. CONCLUSION: Patients have the best survival if all LNmets show tumour regression, even if LNmets are located outside the RT field. Response in LNmets to nCRT is heterogeneous which warrants further studies to better understand underlying mechanisms.


Subject(s)
Chemoradiotherapy , Esophageal Neoplasms , Lymph Nodes , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Humans , Lymph Nodes/pathology , Retrospective Studies , Treatment Outcome
16.
Radiology ; 297(2): 451-458, 2020 11.
Article in English | MEDLINE | ID: mdl-32840472

ABSTRACT

Background Solid components of part-solid nodules (PSNs) at CT are reflective of invasive adenocarcinoma, but studies describing radiomic features of PSNs and the perinodular region are lacking. Purpose To develop and to validate radiomic signatures diagnosing invasive lung adenocarcinoma in PSNs compared with the Brock, clinical-semantic features, and volumetric models. Materials and Methods This retrospective multicenter study (https://ClinicalTrials.gov, NCT03872362) included 291 patients (median age, 60 years; interquartile range, 55-65 years; 191 women) from January 2013 to October 2017 with 297 PSN lung adenocarcinomas split into training (n = 229) and test (n = 68) data sets. Radiomic features were extracted from the different regions (gross tumor volume [GTV], solid, ground-glass, and perinodular). Random-forest models were trained using clinical-semantic, volumetric, and radiomic features, and an online nodule calculator was used to compute the Brock model. Performances of models were evaluated using standard metrics such as area under the curve (AUC), accuracy, and calibration. The integrated discrimination improvement was applied to assess model performance changes after the addition of perinodular features. Results The radiomics model based on ground-glass and solid features yielded an AUC of 0.98 (95% confidence interval [CI]: 0.96, 1.00) on the test data set, which was significantly higher than the Brock (AUC, 0.83 [95% CI: 0.72, 0.94]; P = .007), clinical-semantic (AUC, 0.90 [95% CI: 0.83, 0.98]; P = .03), volumetric GTV (AUC, 0.87 [95% CI: 0.78, 0.96]; P = .008), and radiomics GTV (AUC, 0.88 [95% CI: 0.80, 0.96]; P = .01) models. It also achieved the best accuracy (93% [95% CI: 84%, 98%]). Both this model and the model with added perinodular features showed good calibration, whereas adding perinodular features did not improve the performance (integrated discrimination improvement, -0.02; P = .56). Conclusion Separating ground-glass and solid CT radiomic features of part-solid nodules was useful in diagnosing the invasiveness of lung adenocarcinoma, yielding a better predictive performance than the Brock, clinical-semantic, volumetric, and radiomics gross tumor volume models. Online supplemental material is available for this article. See also the editorial by Nishino in this issue. Published under a CC BY 4.0 license.


Subject(s)
Adenocarcinoma of Lung/diagnostic imaging , Lung Neoplasms/diagnostic imaging , Neoplasm Invasiveness/diagnostic imaging , Solitary Pulmonary Nodule/diagnostic imaging , Tomography, X-Ray Computed , Adenocarcinoma of Lung/pathology , Aged , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness/pathology , Retrospective Studies , Solitary Pulmonary Nodule/pathology
17.
Radiology ; 295(2): 328-338, 2020 05.
Article in English | MEDLINE | ID: mdl-32154773

ABSTRACT

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.


Subject(s)
Biomarkers/analysis , Image Processing, Computer-Assisted/standards , Software , Calibration , Fluorodeoxyglucose F18 , Humans , Lung Neoplasms/diagnostic imaging , Magnetic Resonance Imaging , Phantoms, Imaging , Phenotype , Positron-Emission Tomography , Radiopharmaceuticals , Reproducibility of Results , Sarcoma/diagnostic imaging , Tomography, X-Ray Computed
18.
Eur Respir J ; 56(2)2020 08.
Article in English | MEDLINE | ID: mdl-32616597

ABSTRACT

BACKGROUND: The outbreak of coronavirus disease 2019 (COVID-19) has globally strained medical resources and caused significant mortality. OBJECTIVE: To develop and validate a machine-learning model based on clinical features for severity risk assessment and triage for COVID-19 patients at hospital admission. METHOD: 725 patients were used to train and validate the model. This included a retrospective cohort from Wuhan, China of 299 hospitalised COVID-19 patients from 23 December 2019 to 13 February 2020, and five cohorts with 426 patients from eight centres in China, Italy and Belgium from 20 February 2020 to 21 March 2020. The main outcome was the onset of severe or critical illness during hospitalisation. Model performances were quantified using the area under the receiver operating characteristic curve (AUC) and metrics derived from the confusion matrix. RESULTS: In the retrospective cohort, the median age was 50 years and 137 (45.8%) were male. In the five test cohorts, the median age was 62 years and 236 (55.4%) were male. The model was prospectively validated on five cohorts yielding AUCs ranging from 0.84 to 0.93, with accuracies ranging from 74.4% to 87.5%, sensitivities ranging from 75.0% to 96.9%, and specificities ranging from 55.0% to 88.0%, most of which performed better than the pneumonia severity index. The cut-off values of the low-, medium- and high-risk probabilities were 0.21 and 0.80. The online calculators can be found at www.covid19risk.ai. CONCLUSION: The machine-learning model, nomogram and online calculator might be useful to access the onset of severe and critical illness among COVID-19 patients and triage at hospital admission.


Subject(s)
Coronavirus Infections/diagnosis , Hospital Mortality/trends , Machine Learning , Pneumonia, Viral/diagnosis , Triage/methods , Adult , Age Factors , Aged , Area Under Curve , Belgium , COVID-19 , COVID-19 Testing , China , Clinical Laboratory Techniques , Cohort Studies , Coronavirus Infections/epidemiology , Decision Support Systems, Clinical , Female , Hospitalization/statistics & numerical data , Humans , Internationality , Italy , Male , Middle Aged , Pandemics/statistics & numerical data , Pneumonia, Viral/epidemiology , Predictive Value of Tests , ROC Curve , Reproducibility of Results , Retrospective Studies , Risk Assessment , Severity of Illness Index , Sex Factors , Survival Analysis
19.
BMC Cancer ; 20(1): 557, 2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32539805

ABSTRACT

BACKGROUND: About 50% of non-small cell lung cancer (NSCLC) patients have metastatic disease at initial diagnosis, which limits their treatment options and, consequently, the 5-year survival rate (15%). Immune checkpoint inhibitors (ICI), either alone or in combination with chemotherapy, have become standard of care (SOC) for most good performance status patients. However, most patients will not obtain long-term benefit and new treatment strategies are therefore needed. We previously demonstrated clinical safety of the tumour-selective immunocytokine L19-IL2, consisting of the anti-ED-B scFv L19 antibody coupled to IL2, combined with stereotactic ablative radiotherapy (SABR). METHODS: This investigator-initiated, multicentric, randomised controlled open-label phase II clinical trial will test the hypothesis that the combination of SABR and L19-IL2 increases progression free survival (PFS) in patients with limited metastatic NSCLC. One hundred twenty-six patients will be stratified according to their metastatic load (oligo-metastatic: ≤5 or poly-metastatic: 6 to 10) and randomised to the experimental-arm (E-arm) or the control-arm (C-arm). The C-arm will receive SOC, according to the local protocol. E-arm oligo-metastatic patients will receive SABR to all lesions followed by L19-IL2 therapy; radiotherapy for poly-metastatic patients consists of irradiation of one (symptomatic) to a maximum of 5 lesions (including ICI in both arms if this is the SOC). The accrual period will be 2.5-years, starting after the first centre is initiated and active. Primary endpoint is PFS at 1.5-years based on blinded radiological review, and secondary endpoints are overall survival, toxicity, quality of life and abscopal response. Associative biomarker studies, immune monitoring, CT-based radiomics, stool collection, iRECIST and tumour growth rate will be performed. DISCUSSION: The combination of SABR with or without ICI and the immunocytokine L19-IL2 will be tested as 1st, 2nd or 3rd line treatment in stage IV NSCLC patients in 14 centres located in 6 countries. This bimodal and trimodal treatment approach is based on the direct cytotoxic effect of radiotherapy, the tumour selective immunocytokine L19-IL2, the abscopal effect observed distant from the irradiated metastatic site(s) and the memory effect. The first results are expected end 2023. TRIAL REGISTRATION: ImmunoSABR Protocol Code: NL67629.068.18; EudraCT: 2018-002583-11; Clinicaltrials.gov: NCT03705403; ISRCTN ID: ISRCTN49817477; Date of registration: 03-April-2019.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Chemoradiotherapy/methods , Lung Neoplasms/therapy , Radiosurgery/methods , Recombinant Fusion Proteins/administration & dosage , Adult , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/secondary , Chemoradiotherapy/adverse effects , Clinical Trials, Phase II as Topic , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Multicenter Studies as Topic , Progression-Free Survival , Quality of Life , Radiosurgery/adverse effects , Randomized Controlled Trials as Topic , Recombinant Fusion Proteins/adverse effects , Response Evaluation Criteria in Solid Tumors , Standard of Care
20.
Eur Radiol ; 30(5): 2680-2691, 2020 May.
Article in English | MEDLINE | ID: mdl-32006165

ABSTRACT

OBJECTIVES: Develop a CT-based radiomics model and combine it with frozen section (FS) and clinical data to distinguish invasive adenocarcinomas (IA) from preinvasive lesions/minimally invasive adenocarcinomas (PM). METHODS: This multicenter study cohort of 623 lung adenocarcinomas was split into training (n = 331), testing (n = 143), and external validation dataset (n = 149). Random forest models were built using selected radiomics features, results from FS, lesion volume, clinical and semantic features, and combinations thereof. The area under the receiver operator characteristic curves (AUC) was used to evaluate model performances. The diagnosis accuracy, calibration, and decision curves of models were tested. RESULTS: The radiomics-based model shows good predictive performance and diagnostic accuracy for distinguishing IA from PM, with AUCs of 0.89, 0.89, and 0.88, in the training, testing, and validation datasets, respectively, and with corresponding accuracies of 0.82, 0.79, and 0.85. Adding lesion volume and FS significantly increases the performance of the model with AUCs of 0.96, 0.97, and 0.96, and with accuracies of 0.91, 0.94, and 0.93 in the three datasets. There is no significant difference in AUC between the FS model enriched with radiomics and volume against an FS model enriched with volume alone, while the former has higher accuracy. The model combining all available information shows minor non-significant improvements in AUC and accuracy compared with an FS model enriched with radiomics and volume. CONCLUSIONS: Radiomics signatures are potential biomarkers for the risk of IA, especially in combination with FS, and could help guide surgical strategy for pulmonary nodules patients. KEY POINTS: • A CT-based radiomics model may be a valuable tool for preoperative prediction of invasive adenocarcinoma for patients with pulmonary nodules. • Radiomics combined with frozen sections could help in guiding surgery strategy for patients with pulmonary nodules.


Subject(s)
Adenocarcinoma in Situ/diagnostic imaging , Adenocarcinoma of Lung/diagnostic imaging , Lung Neoplasms/diagnostic imaging , Multiple Pulmonary Nodules/diagnostic imaging , Solitary Pulmonary Nodule/diagnostic imaging , Adenocarcinoma in Situ/pathology , Adenocarcinoma in Situ/surgery , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/surgery , Area Under Curve , Female , Frozen Sections , Humans , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Male , Middle Aged , Multiple Pulmonary Nodules/pathology , Multiple Pulmonary Nodules/surgery , Preoperative Care , ROC Curve , Retrospective Studies , Solitary Pulmonary Nodule/pathology , Tomography, X-Ray Computed/methods
SELECTION OF CITATIONS
SEARCH DETAIL