Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 6756, 2023 10 24.
Article in English | MEDLINE | ID: mdl-37875466

ABSTRACT

High grade serous ovarian carcinoma (HGSOC) is a highly heterogeneous disease that typically presents at an advanced, metastatic state. The multi-scale complexity of HGSOC is a major obstacle to predicting response to neoadjuvant chemotherapy (NACT) and understanding critical determinants of response. Here we present a framework to predict the response of HGSOC patients to NACT integrating baseline clinical, blood-based, and radiomic biomarkers extracted from all primary and metastatic lesions. We use an ensemble machine learning model trained to predict the change in total disease volume using data obtained at diagnosis (n = 72). The model is validated in an internal hold-out cohort (n = 20) and an independent external patient cohort (n = 42). In the external cohort the integrated radiomics model reduces the prediction error by 8% with respect to the clinical model, achieving an AUC of 0.78 for RECIST 1.1 classification compared to 0.47 for the clinical model. Our results emphasize the value of including radiomics data in integrative models of treatment response and provide methods for developing new biomarker-based clinical trials of NACT in HGSOC.


Subject(s)
Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Neoadjuvant Therapy/methods , Biomarkers, Tumor/genetics
2.
Front Oncol ; 12: 868265, 2022.
Article in English | MEDLINE | ID: mdl-35785153

ABSTRACT

Background: Pathological response to neoadjuvant treatment for patients with high-grade serous ovarian carcinoma (HGSOC) is assessed using the chemotherapy response score (CRS) for omental tumor deposits. The main limitation of CRS is that it requires surgical sampling after initial neoadjuvant chemotherapy (NACT) treatment. Earlier and non-invasive response predictors could improve patient stratification. We developed computed tomography (CT) radiomic measures to predict neoadjuvant response before NACT using CRS as a gold standard. Methods: Omental CT-based radiomics models, yielding a simplified fully interpretable radiomic signature, were developed using Elastic Net logistic regression and compared to predictions based on omental tumor volume alone. Models were developed on a single institution cohort of neoadjuvant-treated HGSOC (n = 61; 41% complete response to NCT) and tested on an external test cohort (n = 48; 21% complete response). Results: The performance of the comprehensive radiomics models and the fully interpretable radiomics model was significantly higher than volume-based predictions of response in both the discovery and external test sets when assessed using G-mean (geometric mean of sensitivity and specificity) and NPV, indicating high generalizability and reliability in identifying non-responders when using radiomics. The performance of a fully interpretable model was similar to that of comprehensive radiomics models. Conclusions: CT-based radiomics allows for predicting response to NACT in a timely manner and without the need for abdominal surgery. Adding pre-NACT radiomics to volumetry improved model performance for predictions of response to NACT in HGSOC and was robust to external testing. A radiomic signature based on five robust predictive features provides improved clinical interpretability and may thus facilitate clinical acceptance and application.

3.
Cancers (Basel) ; 14(5)2022 Feb 26.
Article in English | MEDLINE | ID: mdl-35267534

ABSTRACT

The balance between laminin isoforms containing the α5 or the α4 chain in the endothelial basement membrane determines the site of leukocyte diapedesis under inflammatory conditions. Extracellular superoxide dismutase (SOD3) induces laminin α4 expression in tumor blood vessels, which is associated with enhanced intratumor T cell infiltration in primary human cancers. We show now that SOD3 overexpression in neoplastic and endothelial cells (ECs) reduces laminin α5 in tumor blood vessels. SOD3 represses the laminin α5 gene (LAMA5), but LAMA5 expression is not changed in SOD1-overexpressing cells. Transcriptomic analyses revealed SOD3 overexpression to change the transcription of 1682 genes in ECs, with the canonical and non-canonical NF-κB pathways as the major SOD3 targets. Indeed, SOD3 reduced the transcription of well-known NF-κB target genes as well as NF-κB-driven promoter activity in ECs stimulated with tumor necrosis factor (TNF)-α, an NF-κB signaling inducer. SOD3 inhibited the phosphorylation and degradation of IκBα (nuclear factor of the kappa light polypeptide gene enhancer in B-cells inhibitor alpha), an NF-κB inhibitor. Finally, TNF-α was found to be a transcriptional activator of LAMA5 but not of LAMA4; LAMA5 induction was prevented by SOD3. In conclusion, SOD3 is a major regulator of laminin balance in the basement membrane of tumor ECs, with potential implications for immune cell infiltration into tumors.

4.
Eur Radiol ; 31(6): 3765-3772, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33315123

ABSTRACT

PURPOSE: To develop a precision tissue sampling technique that uses computed tomography (CT)-based radiomic tumour habitats for ultrasound (US)-guided targeted biopsies that can be integrated in the clinical workflow of patients with high-grade serous ovarian cancer (HGSOC). METHODS: Six patients with suspected HGSOC scheduled for US-guided biopsy before starting neoadjuvant chemotherapy were included in this prospective study from September 2019 to February 2020. The tumour segmentation was performed manually on the pre-biopsy contrast-enhanced CT scan. Spatial radiomic maps were used to identify tumour areas with similar or distinct radiomic patterns, and tumour habitats were identified using the Gaussian mixture modelling. CT images with superimposed habitat maps were co-registered with US images by means of a landmark-based rigid registration method for US-guided targeted biopsies. The dice similarity coefficient (DSC) was used to assess the tumour-specific CT/US fusion accuracy. RESULTS: We successfully co-registered CT-based radiomic tumour habitats with US images in all patients. The median time between CT scan and biopsy was 21 days (range 7-30 days). The median DSC for tumour-specific CT/US fusion accuracy was 0.53 (range 0.79 to 0.37). The CT/US fusion accuracy was high for the larger pelvic tumours (DSC: 0.76-0.79) while it was lower for the smaller omental metastases (DSC: 0.37-0.53). CONCLUSION: We developed a precision tissue sampling technique that uses radiomic habitats to guide in vivo biopsies using CT/US fusion and that can be seamlessly integrated in the clinical routine for patients with HGSOC. KEY POINTS: • We developed a prevision tissue sampling technique that co-registers CT-based radiomics-based tumour habitats with US images. • The CT/US fusion accuracy was high for the larger pelvic tumours (DSC: 0.76-0.79) while it was lower for the smaller omental metastases (DSC: 0.37-0.53).


Subject(s)
Ovarian Neoplasms , Tomography, X-Ray Computed , Ecosystem , Female , Humans , Ovarian Neoplasms/diagnostic imaging , Prospective Studies , Ultrasonography, Interventional
5.
PLoS One ; 15(11): e0242597, 2020.
Article in English | MEDLINE | ID: mdl-33253194

ABSTRACT

BACKGROUND AND PURPOSE: Few tools are available to predict tumor response to treatment. This retrospective study assesses visual and automatic heterogeneity from 18F-FDG PET images as predictors of response in locally advanced rectal cancer. METHODS: This study included 37 LARC patients who underwent an 18F-FDG PET before their neoadjuvant therapy. One expert segmented the tumor from the PET images. Blinded to the patient´s outcome, two experts established by consensus a visual score for tumor heterogeneity. Metabolic and texture parameters were extracted from the tumor area. Multivariate binary logistic regression with cross-validation was used to estimate the clinical relevance of these features. Area under the ROC Curve (AUC) of each model was evaluated. Histopathological tumor regression grade was the ground-truth. RESULTS: Standard metabolic parameters could discriminate 50.1% of responders (AUC = 0.685). Visual heterogeneity classification showed correct assessment of the response in 75.4% of the sample (AUC = 0.759). Automatic quantitative evaluation of heterogeneity achieved a similar predictive capacity (73.1%, AUC = 0.815). CONCLUSION: A response prediction model in LARC based on tumor heterogeneity (assessed either visually or with automatic texture measurement) shows that texture features may complement the information provided by the metabolic parameters and increase prediction accuracy.


Subject(s)
Fluorodeoxyglucose F18/analysis , Rectal Neoplasms/diagnostic imaging , Rectal Neoplasms/therapy , Aged , Feasibility Studies , Female , Humans , Male , Middle Aged , Neoadjuvant Therapy , Positron-Emission Tomography , Radiotherapy , Rectal Neoplasms/surgery , Treatment Outcome
6.
Insights Imaging ; 11(1): 94, 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32804260

ABSTRACT

BACKGROUND: Ovarian cancer survival rates have not changed in the last 20 years. The majority of cases are High-grade serous ovarian carcinomas (HGSOCs), which are typically diagnosed at an advanced stage with multiple metastatic lesions. Taking biopsies of all sites of disease is infeasible, which challenges the implementation of stratification tools based on molecular profiling. MAIN BODY: In this review, we describe how these challenges might be overcome by integrating quantitative features extracted from medical imaging with the analysis of paired genomic profiles, a combined approach called radiogenomics, to generate virtual biopsies. Radiomic studies have been used to model different imaging phenotypes, and some radiomic signatures have been associated with paired molecular profiles to monitor spatiotemporal changes in the heterogeneity of tumours. We describe different strategies to integrate radiogenomic information in a global and local manner, the latter by targeted sampling of tumour habitats, defined as regions with distinct radiomic phenotypes. CONCLUSION: Linking radiomics and biological correlates in a targeted manner could potentially improve the clinical management of ovarian cancer. Radiogenomic signatures could be used to monitor tumours during the course of therapy, offering additional information for clinical decision making. In summary, radiogenomics may pave the way to virtual biopsies and treatment monitoring tools for integrative tumour analysis.

7.
Comput Biol Med ; 120: 103751, 2020 05.
Article in English | MEDLINE | ID: mdl-32421652

ABSTRACT

BACKGROUND: Cancer typically exhibits genotypic and phenotypic heterogeneity, which can have prognostic significance and influence therapy response. Computed Tomography (CT)-based radiomic approaches calculate quantitative features of tumour heterogeneity at a mesoscopic level, regardless of macroscopic areas of hypo-dense (i.e., cystic/necrotic), hyper-dense (i.e., calcified), or intermediately dense (i.e., soft tissue) portions. METHOD: With the goal of achieving the automated sub-segmentation of these three tissue types, we present here a two-stage computational framework based on unsupervised Fuzzy C-Means Clustering (FCM) techniques. No existing approach has specifically addressed this task so far. Our tissue-specific image sub-segmentation was tested on ovarian cancer (pelvic/ovarian and omental disease) and renal cell carcinoma CT datasets using both overlap-based and distance-based metrics for evaluation. RESULTS: On all tested sub-segmentation tasks, our two-stage segmentation approach outperformed conventional segmentation techniques: fixed multi-thresholding, the Otsu method, and automatic cluster number selection heuristics for the K-means clustering algorithm. In addition, experiments showed that the integration of the spatial information into the FCM algorithm generally achieves more accurate segmentation results, whilst the kernelised FCM versions are not beneficial. The best spatial FCM configuration achieved average Dice similarity coefficient values starting from 81.94±4.76 and 83.43±3.81 for hyper-dense and hypo-dense components, respectively, for the investigated sub-segmentation tasks. CONCLUSIONS: The proposed intelligent framework could be readily integrated into clinical research environments and provides robust tools for future radiomic biomarker validation.


Subject(s)
Algorithms , Fuzzy Logic , Cluster Analysis , Image Processing, Computer-Assisted , Tomography, X-Ray Computed , Tumor Burden
8.
Nat Commun ; 9(1): 575, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29422508

ABSTRACT

One drawback of chemotherapy is poor drug delivery to tumor cells, due in part to hyperpermeability of the tumor vasculature. Extracellular superoxide dismutase (SOD3) is an antioxidant enzyme usually repressed in the tumor milieu. Here we show that specific SOD3 re-expression in tumor-associated endothelial cells (ECs) increases doxorubicin (Doxo) delivery into and chemotherapeutic effect on tumors. Enhanced SOD3 activity fostered perivascular nitric oxide accumulation and reduced vessel leakage by inducing vascular endothelial cadherin (VEC) transcription. SOD3 reduced HIF prolyl hydroxylase domain protein activity, which increased hypoxia-inducible factor-2α (HIF-2α) stability and enhanced its binding to a specific VEC promoter region. EC-specific HIF-2α ablation prevented both the SOD3-mediated increase in VEC transcription and the enhanced Doxo effect. SOD3, VEC, and HIF-2α levels correlated positively in primary colorectal cancers, which suggests a similar interconnection of these proteins in human malignancy.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/metabolism , Doxorubicin/administration & dosage , Endothelial Cells/metabolism , Neoplasms/drug therapy , Superoxide Dismutase/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antineoplastic Agents/administration & dosage , Basic Helix-Loop-Helix Transcription Factors/genetics , Cadherins/genetics , Cadherins/metabolism , Dioxygenases/genetics , Dioxygenases/metabolism , Drug Therapy , Endothelial Cells/drug effects , Female , Humans , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/metabolism , Protein Stability , Superoxide Dismutase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...