Charlotte Robert
Centre national de la recherche scientifique
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Charlotte Robert.
Physics in Medicine and Biology | 2013
Charlotte Robert; George Dedes; G. Battistoni; T.T. Böhlen; Irène Buvat; F. Cerutti; M P W Chin; A. Ferrari; Pierre Gueth; Christopher Kurz; Loïc Lestand; A. Mairani; G. Montarou; R Nicolini; Pablo G. Ortega; Katia Parodi; Y Prezado; P. Sala; David Sarrut; E. Testa
Monte Carlo simulations play a crucial role for in-vivo treatment monitoring based on PET and prompt gamma imaging in proton and carbon-ion therapies. The accuracy of the nuclear fragmentation models implemented in these codes might affect the quality of the treatment verification. In this paper, we investigate the nuclear models implemented in GATE/Geant4 and FLUKA by comparing the angular and energy distributions of secondary particles exiting a homogeneous target of PMMA. Comparison results were restricted to fragmentation of (16)O and (12)C. Despite the very simple target and set-up, substantial discrepancies were observed between the two codes. For instance, the number of high energy (>1 MeV) prompt gammas exiting the target was about twice as large with GATE/Geant4 than with FLUKA both for proton and carbon ion beams. Such differences were not observed for the predicted annihilation photon production yields, for which ratios of 1.09 and 1.20 were obtained between GATE and FLUKA for the proton beam and the carbon ion beam, respectively. For neutrons and protons, discrepancies from 14% (exiting protons-carbon ion beam) to 57% (exiting neutrons-proton beam) have been identified in production yields as well as in the energy spectra for neutrons.
Physics in Medicine and Biology | 2012
Enrica Seravalli; Charlotte Robert; Julia Bauer; Frédéric Stichelbaut; Christopher Kurz; Julien Smeets; C Van Ngoc Ty; Dennis R. Schaart; I Buvat; Katia Parodi; Frank Verhaegen
Positron emission tomography (PET) is a promising tool for monitoring the three-dimensional dose distribution in charged particle radiotherapy. PET imaging during or shortly after proton treatment is based on the detection of annihilation photons following the ß(+)-decay of radionuclides resulting from nuclear reactions in the irradiated tissue. Therapy monitoring is achieved by comparing the measured spatial distribution of irradiation-induced ß(+)-activity with the predicted distribution based on the treatment plan. The accuracy of the calculated distribution depends on the correctness of the computational models, implemented in the employed Monte Carlo (MC) codes that describe the interactions of the charged particle beam with matter and the production of radionuclides and secondary particles. However, no well-established theoretical models exist for predicting the nuclear interactions and so phenomenological models are typically used based on parameters derived from experimental data. Unfortunately, the experimental data presently available are insufficient to validate such phenomenological hadronic interaction models. Hence, a comparison among the models used by the different MC packages is desirable. In this work, starting from a common geometry, we compare the performances of MCNPX, GATE and PHITS MC codes in predicting the amount and spatial distribution of proton-induced activity, at therapeutic energies, to the already experimentally validated PET modelling based on the FLUKA MC code. In particular, we show how the amount of ß(+)-emitters produced in tissue-like media depends on the physics model and cross-sectional data used to describe the proton nuclear interactions, thus calling for future experimental campaigns aiming at supporting improvements of MC modelling for clinical application of PET monitoring.
Physics in Medicine and Biology | 2010
Charlotte Robert; Guillaume Montemont; Véronique Rebuffel; Irène Buvat; Lucie Guerin; Loick Verger
A new gamma-camera architecture named HiSens is presented and evaluated. It consists of a parallel hole collimator, a pixelated CdZnTe (CZT) detector associated with specific electronics for 3D localization and dedicated reconstruction algorithms. To gain in efficiency, a high aperture collimator is used. The spatial resolution is preserved thanks to accurate 3D localization of the interactions inside the detector based on a fine sampling of the CZT detector and on the depth of interaction information. The performance of this architecture is characterized using Monte Carlo simulations in both planar and tomographic modes. Detective quantum efficiency (DQE) computations are then used to optimize the collimator aperture. In planar mode, the simulations show that the fine CZT detector pixelization increases the system sensitivity by 2 compared to a standard Anger camera without loss in spatial resolution. These results are then validated against experimental data. In SPECT, Monte Carlo simulations confirm the merits of the HiSens architecture observed in planar imaging.
Medical Physics | 2011
Charlotte Robert; Guillaume Montemont; Véronique Rebuffel; Loick Verger; Irène Buvat
PURPOSE Small field-of-view CdZnTe (CZT) gamma cameras are increasingly studied for breast lesion detection to complement mammography or ultrasonographic findings. However, in classical collimation configurations, they remain limited by the trade-off between spatial resolution and sensitivity. The HiSens architecture was proposed to overcome these limitations. Using an accurate 3D localization of the interactions inside the detector, this architecture leads to a gain in sensitivity without loss in spatial resolution. In this article, the relevance of the HiSens architecture for planar scintimammography is studied. METHODS A detective quantum efficiency (DQE) computation method is developed and used to optimize the dimensioning of a parallel hole collimator dedicated to scintimammography. Based on the DQE curves, the impact of the collimator-to-detector distance is studied. Two algorithms are proposed to combine data acquired with different collimator-to-detector distances. RESULTS It is shown that CZT detector virtual pixelization increases system sensitivity by 3.3 while preserving a standard LEHR spatial resolution. The introduction of a gap between the CZT detector and the collimator is useful to modulate the DQE curve shape. The combination of data acquired using different gaps in the image formation process leads to enhanced restoration of the frequency content of the images, resulting in image contrast and spatial resolution improvements. CONCLUSIONS Acquisition duration or injected activity could be markedly reduced if the HiSens architecture with an appropriate collimator-detector gap were used.
Oncotarget | 2017
Sylvain Reuzé; Fanny Orlhac; Cyrus Chargari; Christophe Nioche; Elaine Johanna Limkin; François Riet; Alexandre Escande; Christine Haie-Meder; Laurent Dercle; Sebastien Gouy; Irène Buvat; Eric Deutsch; Charlotte Robert
Objectives To identify an imaging signature predicting local recurrence for locally advanced cervical cancer (LACC) treated by chemoradiation and brachytherapy from baseline 18F-FDG PET images, and to evaluate the possibility of gathering images from two different PET scanners in a radiomic study. Methods 118 patients were included retrospectively. Two groups (G1, G2) were defined according to the PET scanner used for image acquisition. Eleven radiomic features were extracted from delineated cervical tumors to evaluate: (i) the predictive value of features for local recurrence of LACC, (ii) their reproducibility as a function of the scanner within a hepatic reference volume, (iii) the impact of voxel size on feature values. Results Eight features were statistically significant predictors of local recurrence in G1 (p < 0.05). The multivariate signature trained in G2 was validated in G1 (AUC=0.76, p<0.001) and identified local recurrence more accurately than SUVmax (p=0.022). Four features were significantly different between G1 and G2 in the liver. Spatial resampling was not sufficient to explain the stratification effect. Conclusion This study showed that radiomic features could predict local recurrence of LACC better than SUVmax. Further investigation is needed before applying a model designed using data from one PET scanner to another.
Frontiers in Oncology | 2017
Elodie Doger de Spéville; Charlotte Robert; Martin Perez-Guevara; Antoine Grigis; Stéphanie Bolle; Clemence Pinaud; Christelle Dufour; A. Beaudré; Virginie Kieffer; Audrey Longaud; Jacques Grill; Dominique Valteau-Couanet; Eric Deutsch; Dimitri Lefkopoulos; Catherine Chiron; Lucie Hertz-Pannier; Marion Noulhiane
Pediatric posterior fossa tumor (PFT) survivors who have been treated with cranial radiation therapy often suffer from cognitive impairments that might relate to IQ decline. Radiotherapy (RT) distinctly affects brain regions involved in different cognitive functions. However, the relative contribution of regional irradiation to the different cognitive impairments still remains unclear. We investigated the relationships between the changes in different cognitive scores and radiation dose distribution in 30 children treated for a PFT. Our exploratory analysis was based on a principal component analysis (PCA) and an ordinary least square regression approach. The use of a PCA was an innovative way to cluster correlated irradiated regions due to similar radiation therapy protocols across patients. Our results suggest an association between working memory decline and a high dose (equivalent uniform dose, EUD) delivered to the orbitofrontal regions, whereas the decline of processing speed seemed more related to EUD in the temporal lobes and posterior fossa. To identify regional effects of RT on cognitive functions may help to propose a rehabilitation program adapted to the risk of cognitive impairment.
Lancet Oncology | 2018
Roger Sun; Elaine Johanna Limkin; Maria Vakalopoulou; Laurent Dercle; Stéphane Champiat; Shan Rong Han; Loic Verlingue; David Brandao; Andrea Lancia; Samy Ammari; Antoine Hollebecque; Jean-Yves Scoazec; Aurélien Marabelle; Christophe Massard; Jean-Charles Soria; Charlotte Robert; Nikos Paragios; Eric Deutsch; Charles Ferté
BACKGROUND Because responses of patients with cancer to immunotherapy can vary in success, innovative predictors of response to treatment are urgently needed to improve treatment outcomes. We aimed to develop and independently validate a radiomics-based biomarker of tumour-infiltrating CD8 cells in patients included in phase 1 trials of anti-programmed cell death protein (PD)-1 or anti-programmed cell death ligand 1 (PD-L1) monotherapy. We also aimed to evaluate the association between the biomarker, and tumour immune phenotype and clinical outcomes of these patients. METHODS In this retrospective multicohort study, we used four independent cohorts of patients with advanced solid tumours to develop and validate a radiomic signature predictive of immunotherapy response by combining contrast-enhanced CT images and RNA-seq genomic data from tumour biopsies to assess CD8 cell tumour infiltration. To develop the radiomic signature of CD8 cells, we used the CT images and RNA sequencing data of 135 patients with advanced solid malignant tumours who had been enrolled into the MOSCATO trial between May 1, 2012, and March 31, 2016, in France (training set). The genomic data, which are based on the CD8B gene, were used to estimate the abundance of CD8 cells in the samples and data were then aligned with the images to generate the radiomic signatures. The concordance of the radiomic signature (primary endpoint) was validated in a Cancer Genome Atlas [TGCA] database dataset including 119 patients who had available baseline preoperative imaging data and corresponding transcriptomic data on June 30, 2017. From 84 input variables used for the machine-learning method (78 radiomic features, five location variables, and one technical variable), a radiomics-based predictor of the CD8 cell expression signature was built by use of machine learning (elastic-net regularised regression method). Two other independent cohorts of patients with advanced solid tumours were used to evaluate this predictor. The immune phenotype internal cohort (n=100), were randomly selected from the Gustave Roussy Cancer Campus database of patient medical records based on previously described, extreme tumour-immune phenotypes: immune-inflamed (with dense CD8 cell infiltration) or immune-desert (with low CD8 cell infiltration), irrespective of treatment delivered; these data were used to analyse the correlation of the immune phenotype with this biomarker. Finally, the immunotherapy-treated dataset (n=137) of patients recruited from Dec 1, 2011, to Jan 31, 2014, at the Gustave Roussy Cancer Campus, who had been treated with anti-PD-1 and anti-PD-L1 monotherapy in phase 1 trials, was used to assess the predictive value of this biomarker in terms of clinical outcome. FINDINGS We developed a radiomic signature for CD8 cells that included eight variables, which was validated with the gene expression signature of CD8 cells in the TCGA dataset (area under the curve [AUC]=0·67; 95% CI 0·57-0·77; p=0·0019). In the cohort with assumed immune phenotypes, the signature was also able to discriminate inflamed tumours from immune-desert tumours (0·76; 0·66-0·86; p<0·0001). In patients treated with anti-PD-1 and PD-L1, a high baseline radiomic score (relative to the median) was associated with a higher proportion of patients who achieved an objective response at 3 months (vs those with progressive disease or stable disease; p=0·049) and a higher proportion of patients who had an objective response (vs those with progressive disease or stable disease; p=0·025) or stable disease (vs those with progressive disease; p=0·013) at 6 months. A high baseline radiomic score was also associated with improved overall survival in univariate (median overall survival 24·3 months in the high radiomic score group, 95% CI 18·63-42·1; vs 11·5 months in the low radiomic score group, 7·98-15·6; hazard ratio 0·58, 95% CI 0·39-0·87; p=0·0081) and multivariate analyses (0·52, 0·35-0·79; p=0·0022). INTERPRETATION The radiomic signature of CD8 cells was validated in three independent cohorts. This imaging predictor provided a promising way to predict the immune phenotype of tumours and to infer clinical outcomes for patients with cancer who had been treated with anti-PD-1 and PD-L1. Our imaging biomarker could be useful in estimating CD8 cell count and predicting clinical outcomes of patients treated with immunotherapy, when validated by further prospective randomised trials. FUNDING Fondation pour la Recherche Médicale, and SIRIC-SOCRATE 2.0, French Society of Radiation Oncology.
Cancer Radiotherapie | 2017
Roger Sun; Elaine Limkin; Laurent Dercle; Sylvain Reuzé; Evangelia I. Zacharaki; Cyrus Chargari; A. Schernberg; Anne-Sophie Dirand; Anthony Alexis; Nikos Paragios; Eric Deutsch; Charles Ferté; Charlotte Robert
The arrival of immunotherapy has profoundly changed the management of multiple cancers, obtaining unexpected tumour responses. However, until now, the majority of patients do not respond to these new treatments. The identification of biomarkers to determine precociously responding patients is a major challenge. Computational medical imaging (also known as radiomics) is a promising and rapidly growing discipline. This new approach consists in the analysis of high-dimensional data extracted from medical imaging, to further describe tumour phenotypes. This approach has the advantages of being non-invasive, capable of evaluating the tumour and its microenvironment in their entirety, thus characterising spatial heterogeneity, and being easily repeatable over time. The end goal of radiomics is to determine imaging biomarkers as decision support tools for clinical practice and to facilitate better understanding of cancer biology, allowing the assessment of the changes throughout the evolution of the disease and the therapeutic sequence. This review will develop the process of computational imaging analysis and present its potential in immuno-oncology.
Biomarkers | 2018
Roger Sun; Elaine Johanna Limkin; Laurent Dercle; Sylvain Reuzé; Stéphane Champiat; David Brandao; Loic Verlingue; Samy Ammari; Sandrine Aspeslagh; Antoine Hollebecque; Christophe Massard; Aurélien Marabelle; Jean-Yves Scoazec; Charlotte Robert; Jean-Charles Soria; Eric Deutsch; Charles Ferté
Background: The discovery of biomarkers identifying responders to immunotherapy is a major challenge. Tumor and peritumoral immune infiltration has been shown to be associated with response to anti-PD-1/PD-L1. The aim of this study was to develop a radiomics-based imaging tool of tumor immune infiltrate and to assess whether such a tool could predict clinical outcomes of patients treated with anti-PD1/PDL1. Methods: A predictive radiomics-based model of tumor-infiltrating CD8+ T cells was trained using data from the head and neck cohort of The Cancer Imaging Archive (HNSC-TCIA). Two cohorts from our institute were used for validation. Contrast-enhanced CTs of 57 patients from the HNSC-TCIA were manually segmented (tumor and surrounding tissue) and 76 radiomics features extracted. A radiomics-based score was build using radiomics features to predict tumor-infiltrating CD8+ T-cells9 abundance, which was estimated using RNA-sequencing data from The Cancer Genome Atlas, and the Microenvironment Cell Populations-counter signature. As a first validation, this signature was applied to an independent cohort of 100 patients for whom the pathologic tumor immune infiltrate was postulated as either favorable (lymphoma, melanoma, lung, bladder, renal, MSI+ cancers, and adenopathy; 70 patients) or unfavorable (adenoid cystic carcinoma, low-grade neuroendocrine tumors, uterine leiomyoma; 30 patients). The signature was then applied on baseline-CTs of a second external cohort of 139 patients prospectively enrolled in anti PD-1/PD-L1 phase 1 trials. The median of the radiomics-based CD8+ score was used to separate patients into two groups (high and low score). Survival was estimated using Cox-proportional hazards model. Results: We developed a radiomics-based CD8+ signature using the six radiomics features that had highest performance on random forest. In the first external cohort, the radiomics-based CD8 T-cells score was associated with the postulated tumor immune infiltrate (Wilcoxon test, P Conclusions: The radiomics-based signature of CD8+ T cells appears as a promising tool to estimate tumor immune infiltrate and to infer the outcome of patients treated with anti-PD-1/PD-L1. Citation Format: Roger Sun, Elaine Johanna Limkin, Laurent Dercle, Sylvain Reuze, Stephane Champiat, David Brandao, Loic Verlingue, Samy Ammari, Sandrine Aspeslagh, Antoine Hollebecque, Christophe Massard, Aurelien Marabelle, Jean-Yves Scoazec, Charlotte Robert, Jean-Charles Soria, Eric Deutsch, Charles Ferte. Prediction of clinical outcomes of cancer patients treated with anti-PD-1/PD-L1 using a radiomics-based imaging score of immune infiltrate [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26-30; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2018;17(1 Suppl):Abstract nr A051.
Medical Physics | 2014
David Sarrut; Manuel Bardies; Nicolas Boussion; N. Freud; Sébastien Jan; J.M. Létang; George Loudos; Lydia Maigne; Sara Marcatili; Thibault Mauxion; Panagiotis Papadimitroulas; Yann Perrot; U. Pietrzyk; Charlotte Robert; Dennis R. Schaart; Dimitris Visvikis; Irène Buvat