Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
NPJ Digit Med ; 1: 67, 2018.
Article in English | MEDLINE | ID: mdl-31304344

ABSTRACT

Physiological signals have shown to be reliable indicators of stress in laboratory studies, yet large-scale ambulatory validation is lacking. We present a large-scale cross-sectional study for ambulatory stress detection, consisting of 1002 subjects, containing subjects' demographics, baseline psychological information, and five consecutive days of free-living physiological and contextual measurements, collected through wearable devices and smartphones. This dataset represents a healthy population, showing associations between wearable physiological signals and self-reported daily-life stress. Using a data-driven approach, we identified digital phenotypes characterized by self-reported poor health indicators and high depression, anxiety and stress scores that are associated with blunted physiological responses to stress. These results emphasize the need for large-scale collections of multi-sensor data, to build personalized stress models for precision medicine.

2.
Elife ; 62017 07 21.
Article in English | MEDLINE | ID: mdl-28731408

ABSTRACT

Medical imaging can visualize characteristics of human cancer noninvasively. Radiomics is an emerging field that translates these medical images into quantitative data to enable phenotypic profiling of tumors. While radiomics has been associated with several clinical endpoints, the complex relationships of radiomics, clinical factors, and tumor biology are largely unknown. To this end, we analyzed two independent cohorts of respectively 262 North American and 89 European patients with lung cancer, and consistently identified previously undescribed associations between radiomic imaging features, molecular pathways, and clinical factors. In particular, we found a relationship between imaging features, immune response, inflammation, and survival, which was further validated by immunohistochemical staining. Moreover, a number of imaging features showed predictive value for specific pathways; for example, intra-tumor heterogeneity features predicted activity of RNA polymerase transcription (AUC = 0.62, p=0.03) and intensity dispersion was predictive of the autodegration pathway of a ubiquitin ligase (AUC = 0.69, p<10-4). Finally, we observed that prognostic biomarkers performed highest when combining radiomic, genetic, and clinical information (CI = 0.73, p<10-9) indicating complementary value of these data. In conclusion, we demonstrate that radiomic approaches permit noninvasive assessment of both molecular and clinical characteristics of tumors, and therefore have the potential to advance clinical decision-making by systematically analyzing standard-of-care medical images.


Subject(s)
Diagnostic Imaging/methods , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/radiotherapy , Phenotype , Radiometry/methods , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/pathology , Adenocarcinoma/radiotherapy , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/radiotherapy , Clinical Decision-Making , Female , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Male , Prognosis , Tomography, X-Ray Computed/methods
3.
Cancer Res ; 77(14): 3922-3930, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28566328

ABSTRACT

Tumors are characterized by somatic mutations that drive biological processes ultimately reflected in tumor phenotype. With regard to radiographic phenotypes, generally unconnected through present understanding to the presence of specific mutations, artificial intelligence methods can automatically quantify phenotypic characters by using predefined, engineered algorithms or automatic deep-learning methods, a process also known as radiomics. Here we demonstrate how imaging phenotypes can be connected to somatic mutations through an integrated analysis of independent datasets of 763 lung adenocarcinoma patients with somatic mutation testing and engineered CT image analytics. We developed radiomic signatures capable of distinguishing between tumor genotypes in a discovery cohort (n = 353) and verified them in an independent validation cohort (n = 352). All radiomic signatures significantly outperformed conventional radiographic predictors (tumor volume and maximum diameter). We found a radiomic signature related to radiographic heterogeneity that successfully discriminated between EGFR+ and EGFR- cases (AUC = 0.69). Combining this signature with a clinical model of EGFR status (AUC = 0.70) significantly improved prediction accuracy (AUC = 0.75). The highest performing signature was capable of distinguishing between EGFR+ and KRAS+ tumors (AUC = 0.80) and, when combined with a clinical model (AUC = 0.81), substantially improved its performance (AUC = 0.86). A KRAS+/KRAS- radiomic signature also showed significant albeit lower performance (AUC = 0.63) and did not improve the accuracy of a clinical predictor of KRAS status. Our results argue that somatic mutations drive distinct radiographic phenotypes that can be predicted by radiomics. This work has implications for the use of imaging-based biomarkers in the clinic, as applied noninvasively, repeatedly, and at low cost. Cancer Res; 77(14); 3922-30. ©2017 AACR.


Subject(s)
Adenocarcinoma/diagnostic imaging , Adenocarcinoma/genetics , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/genetics , Mutation , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Cohort Studies , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Phenotype , Proto-Oncogene Proteins p21(ras)/biosynthesis , Proto-Oncogene Proteins p21(ras)/genetics , Tomography, X-Ray Computed
4.
Oncotarget ; 7(24): 37288-37296, 2016 Jun 14.
Article in English | MEDLINE | ID: mdl-27095578

ABSTRACT

BACKGROUND AND PURPOSE: To improve quality and personalization of oncology health care, decision aid tools are needed to advise physicians and patients. The aim of this work is to demonstrate the clinical relevance of a survival prediction model as a first step to multi institutional rapid learning and compare this to a clinical trial dataset. MATERIALS AND METHODS: Data extraction and mining tools were used to collect uncurated input parameters from Illawarra Cancer Care Centre's (clinical cohort) oncology information system. Prognosis categories previously established from the Maastricht Radiation Oncology (training cohort) dataset, were applied to the clinical cohort and the radiotherapy only arm of the RTOG-9111 (trial cohort). RESULTS: Data mining identified 125 laryngeal carcinoma patients, ending up with 52 patients in the clinical cohort who were eligible to be evaluated by the model to predict 2-year survival and 177 for the trial cohort. The model was able to classify patients and predict survival in the clinical cohort, but for the trial cohort it failed to do so. CONCLUSIONS: The technical infrastructure and model is able to support the prognosis prediction of laryngeal carcinoma patients in a clinical cohort. The model does not perform well for the highly selective patient population in the trial cohort.


Subject(s)
Clinical Decision-Making/methods , Decision Support Techniques , Laryngeal Neoplasms/mortality , Cohort Studies , Datasets as Topic , Female , Humans , Kaplan-Meier Estimate , Laryngeal Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis
5.
Sci Rep ; 5: 16822, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26576732

ABSTRACT

Reproducible definition and quantification of imaging biomarkers is essential. We evaluated a fully automatic MR-based segmentation method by comparing it to manually defined sub-volumes by experienced radiologists in the TCGA-GBM dataset, in terms of sub-volume prognosis and association with VASARI features. MRI sets of 109 GBM patients were downloaded from the Cancer Imaging archive. GBM sub-compartments were defined manually and automatically using the Brain Tumor Image Analysis (BraTumIA). Spearman's correlation was used to evaluate the agreement with VASARI features. Prognostic significance was assessed using the C-index. Auto-segmented sub-volumes showed moderate to high agreement with manually delineated volumes (range (r): 0.4 - 0.86). Also, the auto and manual volumes showed similar correlation with VASARI features (auto r = 0.35, 0.43 and 0.36; manual r = 0.17, 0.67, 0.41, for contrast-enhancing, necrosis and edema, respectively). The auto-segmented contrast-enhancing volume and post-contrast abnormal volume showed the highest AUC (0.66, CI: 0.55-0.77 and 0.65, CI: 0.54-0.76), comparable to manually defined volumes (0.64, CI: 0.53-0.75 and 0.63, CI: 0.52-0.74, respectively). BraTumIA and manual tumor sub-compartments showed comparable performance in terms of prognosis and correlation with VASARI features. This method can enable more reproducible definition and quantification of imaging based biomarkers and has potential in high-throughput medical imaging research.


Subject(s)
Datasets as Topic , Glioblastoma/diagnosis , Image Interpretation, Computer-Assisted , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Medical Informatics , Glioblastoma/mortality , Humans , Image Interpretation, Computer-Assisted/methods , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Medical Informatics/methods , Prognosis , Reproducibility of Results , Software , Tumor Burden
6.
Sci Rep ; 5: 11044, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-26251068

ABSTRACT

Radiomics provides a comprehensive quantification of tumor phenotypes by extracting and mining large number of quantitative image features. To reduce the redundancy and compare the prognostic characteristics of radiomic features across cancer types, we investigated cancer-specific radiomic feature clusters in four independent Lung and Head &Neck (H) cancer cohorts (in total 878 patients). Radiomic features were extracted from the pre-treatment computed tomography (CT) images. Consensus clustering resulted in eleven and thirteen stable radiomic feature clusters for Lung and H cancer, respectively. These clusters were validated in independent external validation cohorts using rand statistic (Lung RS = 0.92, p < 0.001, H RS = 0.92, p < 0.001). Our analysis indicated both common as well as cancer-specific clustering and clinical associations of radiomic features. Strongest associations with clinical parameters: Prognosis Lung CI = 0.60 ± 0.01, Prognosis H CI = 0.68 ± 0.01; Lung histology AUC = 0.56 ± 0.03, Lung stage AUC = 0.61 ± 0.01, H HPV AUC = 0.58 ± 0.03, H stage AUC = 0.77 ± 0.02. Full utilization of these cancer-specific characteristics of image features may further improve radiomic biomarkers, providing a non-invasive way of quantifying and monitoring tumor phenotypic characteristics in clinical practice.


Subject(s)
Head and Neck Neoplasms/pathology , Lung Neoplasms/pathology , Biomarkers, Tumor/metabolism , Head and Neck Neoplasms/metabolism , Humans , Lung Neoplasms/metabolism , Prognosis , Tomography, X-Ray Computed/methods
7.
Radiother Oncol ; 114(3): 345-50, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25746350

ABSTRACT

BACKGROUND AND PURPOSE: Radiomics provides opportunities to quantify the tumor phenotype non-invasively by applying a large number of quantitative imaging features. This study evaluates computed-tomography (CT) radiomic features for their capability to predict distant metastasis (DM) for lung adenocarcinoma patients. MATERIAL AND METHODS: We included two datasets: 98 patients for discovery and 84 for validation. The phenotype of the primary tumor was quantified on pre-treatment CT-scans using 635 radiomic features. Univariate and multivariate analysis was performed to evaluate radiomics performance using the concordance index (CI). RESULTS: Thirty-five radiomic features were found to be prognostic (CI>0.60, FDR<5%) for DM and twelve for survival. It is noteworthy that tumor volume was only moderately prognostic for DM (CI=0.55, p-value=2.77×10(-5)) in the discovery cohort. A radiomic-signature had strong power for predicting DM in the independent validation dataset (CI=0.61, p-value=1.79×10(-17)). Adding this radiomic-signature to a clinical model resulted in a significant improvement of predicting DM in the validation dataset (p-value=1.56×10(-11)). CONCLUSIONS: Although only basic metrics are routinely quantified, this study shows that radiomic features capturing detailed information of the tumor phenotype can be used as a prognostic biomarker for clinically-relevant factors such as DM. Moreover, the radiomic-signature provided additional information to clinical data.


Subject(s)
Adenocarcinoma/diagnostic imaging , Lung Neoplasms/diagnostic imaging , Adenocarcinoma of Lung , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Prognosis , Tomography, X-Ray Computed/methods , Tumor Burden
8.
Radiother Oncol ; 113(3): 324-30, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25443497

ABSTRACT

PURPOSE: Due to the established role of the human papillomavirus (HPV), the optimal treatment for oropharyngeal carcinoma is currently under debate. We evaluated the most important determinants of treatment outcome to develop a multifactorial predictive model that could provide individualized predictions of treatment outcome in oropharyngeal carcinoma patients. METHODS: We analyzed the association between clinico-pathological factors and overall and progression-free survival in 168 OPSCC patients treated with curative radiotherapy or concurrent chemo-radiation. A multivariate model was validated in an external dataset of 189 patients and compared to the TNM staging system. This nomogram will be made publicly available at www.predictcancer.org. RESULTS: Predictors of unfavorable outcomes were negative HPV-status, moderate to severe comorbidity, T3-T4 classification, N2b-N3 stage, male gender, lower hemoglobin levels and smoking history of more than 30 pack years. Prediction of overall survival using the multi-parameter model yielded a C-index of 0.82 (95% CI, 0.76-0.88). Validation in an independent dataset yielded a C-index of 0.73 (95% CI, 0.66-0.79. For progression-free survival, the model's C-index was 0.80 (95% CI, 0.76-0.88), with a validation C-index of 0.67, (95% CI, 0.59-0.74). Stratification of model estimated probabilities showed statistically different prognosis groups in both datasets (p<0.001). CONCLUSION: This nomogram was superior to TNM classification or HPV status alone in an independent validation dataset for prediction of overall and progression-free survival in OPSCC patients, assigning patients to distinct prognosis groups. These individualized predictions could be used to stratify patients for treatment de-escalation trials.


Subject(s)
Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Chemoradiotherapy/methods , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/therapy , Nomograms , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/virology , Disease-Free Survival , Female , Head and Neck Neoplasms/virology , Humans , Male , Middle Aged , Neoplasm Staging , Oropharyngeal Neoplasms/virology , Papillomaviridae/isolation & purification , Polymerase Chain Reaction/methods , Predictive Value of Tests , Prognosis , Reproducibility of Results , Severity of Illness Index , Sex Factors , Squamous Cell Carcinoma of Head and Neck , Treatment Outcome
9.
Cancer Epidemiol ; 38(5): 591-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25127693

ABSTRACT

INTRODUCTION: This study aimed to investigate the effect of genetic polymorphisms in miRNA sequences, miRNA target genes and miRNA processing genes as additional biomarkers to HPV for prognosis in oropharyngeal squamous cell carcinoma (OPSCC) patients. Secondarily, the prevalence of HPV-associated OPSCC in a European cohort was mapped. METHODS: OPSCC patients (n=122) were genotyped for ten genetic polymorphisms in pre-miRNAs (pre-mir-146a, pre-mir-196a2), in miRNA biosynthesis genes (Drosha, XPO5) and in miRNA target genes (KRAS, SMC1B). HPV status was assessed by p16 immunohistochemistry (IHC) and high-risk HPV in situ hybridization (ISH) or by p16 IHC and PCR followed by enzyme-immunoassay (EIA). Overall and disease specific survival were analysed using Kaplan-Meier plots (log-rank test). Cox proportional hazard model was used to calculate hazard ratios (HR). RESULTS: The overall HPV prevalence rate in our Belgian/Dutch cohort was 27.9%. Patients with HPV(+) tumours had a better 5-years overall survival (78% vs. 46%, p=0.001) and a better 5-years disease specific survival (90% vs. 70%, p=0.016) compared to patients with HPV(-) tumours. In multivariate Cox analysis including clinical, treatment and genetic parameters, HPV negativity (HR=3.89, p=0.005), advanced T-stage (HR=1.81, p=0.050), advanced N-stage (HR=5.86, p=0.001) and >10 pack-years of smoking (HR=3.45, p=0.012) were significantly associated with reduced overall survival. The variant G-allele of the KRAS-LCS6 polymorphism was significantly associated with a better overall survival (HR=0.40, p=0.031). CONCLUSIONS: Our results demonstrate that OPSCC patients with the KRAS-LCS6 variant have a better outcome and suggest that this variant may be used as a prognostic biomarker for OPSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , MicroRNAs/genetics , Oropharyngeal Neoplasms/pathology , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , 3' Untranslated Regions/genetics , Adult , Aged , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Female , Follow-Up Studies , Genotype , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/virology , Papillomavirus Infections/complications , Papillomavirus Infections/epidemiology , Polymorphism, Genetic , Prevalence , Prognosis , Proportional Hazards Models , Proto-Oncogene Proteins p21(ras) , Survival Rate
10.
PLoS One ; 9(7): e102107, 2014.
Article in English | MEDLINE | ID: mdl-25025374

ABSTRACT

Due to advances in the acquisition and analysis of medical imaging, it is currently possible to quantify the tumor phenotype. The emerging field of Radiomics addresses this issue by converting medical images into minable data by extracting a large number of quantitative imaging features. One of the main challenges of Radiomics is tumor segmentation. Where manual delineation is time consuming and prone to inter-observer variability, it has been shown that semi-automated approaches are fast and reduce inter-observer variability. In this study, a semiautomatic region growing volumetric segmentation algorithm, implemented in the free and publicly available 3D-Slicer platform, was investigated in terms of its robustness for quantitative imaging feature extraction. Fifty-six 3D-radiomic features, quantifying phenotypic differences based on tumor intensity, shape and texture, were extracted from the computed tomography images of twenty lung cancer patients. These radiomic features were derived from the 3D-tumor volumes defined by three independent observers twice using 3D-Slicer, and compared to manual slice-by-slice delineations of five independent physicians in terms of intra-class correlation coefficient (ICC) and feature range. Radiomic features extracted from 3D-Slicer segmentations had significantly higher reproducibility (ICC = 0.85±0.15, p = 0.0009) compared to the features extracted from the manual segmentations (ICC = 0.77±0.17). Furthermore, we found that features extracted from 3D-Slicer segmentations were more robust, as the range was significantly smaller across observers (p = 3.819e-07), and overlapping with the feature ranges extracted from manual contouring (boundary lower: p = 0.007, higher: p = 5.863e-06). Our results show that 3D-Slicer segmented tumor volumes provide a better alternative to the manual delineation for feature quantification, as they yield more reproducible imaging descriptors. Therefore, 3D-Slicer can be employed for quantitative image feature extraction and image data mining research in large patient cohorts.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnosis , Diagnostic Imaging/methods , Image Processing, Computer-Assisted , Imaging, Three-Dimensional/methods , Lung Neoplasms/diagnosis , Pattern Recognition, Automated , Humans , Observer Variation , Positron-Emission Tomography , Reproducibility of Results , Tomography, X-Ray Computed/methods
11.
Nat Rev Clin Oncol ; 10(1): 27-40, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23165123

ABSTRACT

With the emergence of individualized medicine and the increasing amount and complexity of available medical data, a growing need exists for the development of clinical decision-support systems based on prediction models of treatment outcome. In radiation oncology, these models combine both predictive and prognostic data factors from clinical, imaging, molecular and other sources to achieve the highest accuracy to predict tumour response and follow-up event rates. In this Review, we provide an overview of the factors that are correlated with outcome-including survival, recurrence patterns and toxicity-in radiation oncology and discuss the methodology behind the development of prediction models, which is a multistage process. Even after initial development and clinical introduction, a truly useful predictive model will be continuously re-evaluated on different patient datasets from different regions to ensure its population-specific strength. In the future, validated decision-support systems will be fully integrated in the clinic, with data and knowledge being shared in a standardized, instant and global manner.


Subject(s)
Decision Support Systems, Clinical , Models, Theoretical , Neoplasms/radiotherapy , Precision Medicine , Radiation Oncology , Humans , Neoplasms/mortality , Treatment Outcome
12.
Radiother Oncol ; 105(2): 167-73, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23157978

ABSTRACT

PURPOSE: To assess the clinical relevance of a semiautomatic CT-based ensemble segmentation method, by comparing it to pathology and to CT/PET manual delineations by five independent radiation oncologists in non-small cell lung cancer (NSCLC). MATERIALS AND METHODS: For 20 NSCLC patients (stages Ib-IIIb) the primary tumor was delineated manually on CT/PET scans by five independent radiation oncologists and segmented using a CT based semi-automatic tool. Tumor volume and overlap fractions between manual and semiautomatic-segmented volumes were compared. All measurements were correlated with the maximal diameter on macroscopic examination of the surgical specimen. Imaging data are available on www.cancerdata.org. RESULTS: High overlap fractions were observed between the semi-automatically segmented volumes and the intersection (92.5±9.0, mean±SD) and union (94.2±6.8) of the manual delineations. No statistically significant differences in tumor volume were observed between the semiautomatic segmentation (71.4±83.2 cm(3), mean±SD) and manual delineations (81.9±94.1 cm(3); p=0.57). The maximal tumor diameter of the semiautomatic-segmented tumor correlated strongly with the macroscopic diameter of the primary tumor (r=0.96). CONCLUSIONS: Semiautomatic segmentation of the primary tumor on CT demonstrated high agreement with CT/PET manual delineations and strongly correlated with the macroscopic diameter considered as the "gold standard". This method may be used routinely in clinical practice and could be employed as a starting point for treatment planning, target definition in multi-center clinical trials or for high throughput data mining research. This method is particularly suitable for peripherally located tumors.


Subject(s)
Lung Neoplasms/diagnostic imaging , Tomography, X-Ray Computed , Algorithms , Humans , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Multimodal Imaging , Positron-Emission Tomography
13.
Eur J Cancer ; 48(4): 441-6, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22257792

ABSTRACT

Solid cancers are spatially and temporally heterogeneous. This limits the use of invasive biopsy based molecular assays but gives huge potential for medical imaging, which has the ability to capture intra-tumoural heterogeneity in a non-invasive way. During the past decades, medical imaging innovations with new hardware, new imaging agents and standardised protocols, allows the field to move towards quantitative imaging. Therefore, also the development of automated and reproducible analysis methodologies to extract more information from image-based features is a requirement. Radiomics--the high-throughput extraction of large amounts of image features from radiographic images--addresses this problem and is one of the approaches that hold great promises but need further validation in multi-centric settings and in the laboratory.


Subject(s)
Diagnostic Imaging , High-Throughput Screening Assays/methods , Image Processing, Computer-Assisted , Radioactive Tracers , Radiometry/statistics & numerical data , Algorithms , Diagnostic Imaging/methods , Diagnostic Imaging/statistics & numerical data , Diagnostic Imaging/trends , Genomics/methods , Humans , Image Processing, Computer-Assisted/methods , Image Processing, Computer-Assisted/statistics & numerical data , Models, Biological , Pattern Recognition, Automated/methods , Proteomics/methods , Radiometry/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...