Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where T Coroller is active.

Publication


Featured researches published by T Coroller.


Radiotherapy and Oncology | 2015

CT-based radiomic signature predicts distant metastasis in lung adenocarcinoma

T Coroller; Patrick Grossmann; Ying Hou; Emmanuel Rios Velazquez; R. Leijenaar; Gretchen Hermann; Philippe Lambin; Benjamin Haibe-Kains; Raymond H. Mak; Hugo J.W.L. Aerts

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.


Radiotherapy and Oncology | 2016

Radiomic phenotype features predict pathological response in non-small cell lung cancer.

T Coroller; Vishesh Agrawal; Vivek Narayan; Ying Hou; Patrick Grossmann; Stephanie W. Lee; Raymond H. Mak; Hugo J.W.L. Aerts

BACKGROUND AND PURPOSE Radiomics can quantify tumor phenotype characteristics non-invasively by applying advanced imaging feature algorithms. In this study we assessed if pre-treatment radiomics data are able to predict pathological response after neoadjuvant chemoradiation in patients with locally advanced non-small cell lung cancer (NSCLC). MATERIALS AND METHODS 127 NSCLC patients were included in this study. Fifteen radiomic features selected based on stability and variance were evaluated for its power to predict pathological response. Predictive power was evaluated using area under the curve (AUC). Conventional imaging features (tumor volume and diameter) were used for comparison. RESULTS Seven features were predictive for pathologic gross residual disease (AUC>0.6, p-value<0.05), and one for pathologic complete response (AUC=0.63, p-value=0.01). No conventional imaging features were predictive (range AUC=0.51-0.59, p-value>0.05). Tumors that did not respond well to neoadjuvant chemoradiation were more likely to present a rounder shape (spherical disproportionality, AUC=0.63, p-value=0.009) and heterogeneous texture (LoG 5mm 3D - GLCM entropy, AUC=0.61, p-value=0.03). CONCLUSION We identified predictive radiomic features for pathological response, although no conventional features were significantly predictive. This study demonstrates that radiomics can provide valuable clinical information, and performed better than conventional imaging features.


Radiotherapy and Oncology | 2016

CT-based radiomic analysis of stereotactic body radiation therapy patients with lung cancer

Elizabeth Huynh; T Coroller; Vivek Narayan; Vishesh Agrawal; Ying Hou; John Romano; I. Franco; Raymond H. Mak; Hugo J.W.L. Aerts

BACKGROUND Radiomics uses a large number of quantitative imaging features that describe the tumor phenotype to develop imaging biomarkers for clinical outcomes. Radiomic analysis of pre-treatment computed-tomography (CT) scans was investigated to identify imaging predictors of clinical outcomes in early stage non-small cell lung cancer (NSCLC) patients treated with stereotactic body radiation therapy (SBRT). MATERIALS AND METHODS CT images of 113 stage I-II NSCLC patients treated with SBRT were analyzed. Twelve radiomic features were selected based on stability and variance. The association of features with clinical outcomes and their prognostic value (using the concordance index (CI)) was evaluated. Radiomic features were compared with conventional imaging metrics (tumor volume and diameter) and clinical parameters. RESULTS Overall survival was associated with two conventional features (volume and diameter) and two radiomic features (LoG 3D run low gray level short run emphasis and stats median). One radiomic feature (Wavelet LLH stats range) was significantly prognostic for distant metastasis (CI=0.67, q-value<0.1), while none of the conventional and clinical parameters were. Three conventional and four radiomic features were prognostic for overall survival. CONCLUSION This exploratory analysis demonstrates that radiomic features have potential to be prognostic for some outcomes that conventional imaging metrics cannot predict in SBRT patients.


Cancer Research | 2017

Somatic mutations drive distinct imaging phenotypes in lung cancer

Emmanuel Rios Velazquez; Chintan Parmar; Ying Liu; T Coroller; Gisele P. Cruz; Olya Stringfield; Zhaoxiang Ye; G. Mike Makrigiorgos; Fiona M. Fennessy; Raymond H. Mak; Robert J. Gillies; John Quackenbush; Hugo J.W.L. Aerts

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.


The Journal of Nuclear Medicine | 2017

Associations between somatic mutations and metabolic imaging phenotypes in non-small cell lung cancer

Stephen Yip; John Kim; T Coroller; Chintan Parmar; Rios Velazquez E; Elizabeth Huynh; Raymond H. Mak; Hugo J.W.L. Aerts

PET-based radiomics have been used to noninvasively quantify the metabolic tumor phenotypes; however, little is known about the relationship between these phenotypes and underlying somatic mutations. This study assessed the association and predictive power of 18F-FDG PET–based radiomic features for somatic mutations in non–small cell lung cancer patients. Methods: Three hundred forty-eight non–small cell lung cancer patients underwent diagnostic 18F-FDG PET scans and were tested for genetic mutations. Thirteen percent (44/348) and 28% (96/348) of patients were found to harbor epidermal growth factor receptor (EGFR) or Kristen rat sarcoma viral (KRAS) mutations, respectively. We evaluated 21 imaging features: 19 independent radiomic features quantifying phenotypic traits and 2 conventional features (metabolic tumor volume and maximum SUV). The association between imaging features and mutation status (e.g., EGFR-positive [EGFR+] vs. EGFR-negative) was assessed using the Wilcoxon rank-sum test. The ability of each imaging feature to predict mutation status was evaluated by the area under the receiver operating curve (AUC) and its significance was compared with a random guess (AUC = 0.5) using the Noether test. All P values were corrected for multiple hypothesis testing by controlling the false-discovery rate (FDRWilcoxon, FDRNoether) with a significance threshold of 10%. Results: Eight radiomic features and both conventional features were significantly associated with EGFR mutation status (FDRWilcoxon = 0.01–0.10). One radiomic feature (normalized inverse difference moment) outperformed all other features in predicting EGFR mutation status (EGFR+ vs. EGFR-negative, AUC = 0.67, FDRNoether = 0.0032), as well as differentiating between KRAS-positive and EGFR+ (AUC = 0.65, FDRNoether = 0.05). None of the features was associated with or predictive of KRAS mutation status (KRAS-positive vs. KRAS-negative, AUC = 0.50–0.54). Conclusion: Our results indicate that EGFR mutations may drive different metabolic tumor phenotypes that are captured in PET images, whereas KRAS-mutated tumors do not. This proof-of-concept study sheds light on genotype–phenotype interactions, using radiomics to capture and describe the phenotype, and may have potential for developing noninvasive imaging biomarkers for somatic mutations.


PLOS ONE | 2014

Low Incidence of Chest Wall Pain with a Risk-Adapted Lung Stereotactic Body Radiation Therapy Approach Using Three or Five Fractions Based on Chest Wall Dosimetry

T Coroller; Raymond H. Mak; John Lewis; Elizabeth H. Baldini; Aileen B. Chen; Yolonda L. Colson; F Hacker; Gretchen Hermann; David Kozono; E Mannarino; Christina Molodowitch; Jon O. Wee; David J. Sher; Joseph H. Killoran

Purpose To examine the frequency and potential of dose-volume predictors for chest wall (CW) toxicity (pain and/or rib fracture) for patients receiving lung stereotactic body radiotherapy (SBRT) using treatment planning methods to minimize CW dose and a risk-adapted fractionation scheme. Methods We reviewed data from 72 treatment plans, from 69 lung SBRT patients with at least one year of follow-up or CW toxicity, who were treated at our center between 2010 and 2013. Treatment plans were optimized to reduce CW dose and patients received a risk-adapted fractionation of 18 Gy×3 fractions (54 Gy total) if the CW V30 was less than 30 mL or 10–12 Gy×5 fractions (50–60 Gy total) otherwise. The association between CW toxicity and patient characteristics, treatment parameters and dose metrics, including biologically equivalent dose, were analyzed using logistic regression. Results With a median follow-up of 20 months, 6 (8.3%) patients developed CW pain including three (4.2%) grade 1, two (2.8%) grade 2 and one (1.4%) grade 3. Five (6.9%) patients developed rib fractures, one of which was symptomatic. No significant associations between CW toxicity and patient and dosimetric variables were identified on univariate nor multivariate analysis. Conclusions Optimization of treatment plans to reduce CW dose and a risk-adapted fractionation strategy of three or five fractions based on the CW V30 resulted in a low incidence of CW toxicity. Under these conditions, none of the patient characteristics or dose metrics we examined appeared to be predictive of CW pain.


Journal of Thoracic Oncology | 2017

Radiomic-Based Pathological Response Prediction from Primary Tumors and Lymph Nodes in NSCLC

T Coroller; Vishesh Agrawal; Elizabeth Huynh; Vivek Narayan; Stephanie W. Lee; Raymond H. Mak; Hugo J.W.L. Aerts

Introduction: Noninvasive biomarkers that capture the total tumor burden could provide important complementary information for precision medicine to aid clinical decision making. We investigated the value of radiomic data extracted from pretreatment computed tomography images of the primary tumor and lymph nodes in predicting pathological response after neoadjuvant chemoradiation before surgery. Methods: A total of 85 patients with resectable locally advanced (stage II–III) NSCLC (median age 60.3 years, 65% female) treated from 2003 to 2013 were included in this institutional review board–approved study. Radiomics analysis was performed on 85 primary tumors and 178 lymph nodes to discriminate between pathological complete response (pCR) and gross residual disease (GRD). Twenty nonredundant and stable features (10 from each site) were evaluated by using the area under the curve (AUC) (all p values were corrected for multiple hypothesis testing). Classification performance of each feature set was evaluated by random forest and nested cross validation. Results: Three radiomic features (describing primary tumor sphericity and lymph node homogeneity) were significantly predictive of pCR with similar performances (all AUC = 0.67, p < 0.05). Two features (quantifying lymph node homogeneity) were predictive of GRD (AUC range 0.72–0.75, p < 0.05) and performed significantly better than the primary features (AUC = 0.62). Multivariate analysis showed that for pCR, the radiomic features set alone had the best‐performing classification (median AUC = 0.68). Furthermore, for GRD classification, the combination of radiomic and clinical data significantly outperformed all other feature sets (median AUC = 0.73). Conclusion: Lymph node phenotypic information was significantly predictive for pathological response and showed higher classification performance than radiomic features obtained from the primary tumor.


PLOS ONE | 2017

Associations of Radiomic Data Extracted from Static and Respiratory-Gated CT Scans with Disease Recurrence in Lung Cancer Patients Treated with SBRT.

Elizabeth Huynh; T Coroller; Vivek Narayan; Vishesh Agrawal; John Romano; I. Franco; Chintan Parmar; Ying Hou; Raymond H. Mak; Hugo J.W.L. Aerts

Radiomics aims to quantitatively capture the complex tumor phenotype contained in medical images to associate them with clinical outcomes. This study investigates the impact of different types of computed tomography (CT) images on the prognostic performance of radiomic features for disease recurrence in early stage non-small cell lung cancer (NSCLC) patients treated with stereotactic body radiation therapy (SBRT). 112 early stage NSCLC patients treated with SBRT that had static free breathing (FB) and average intensity projection (AIP) images were analyzed. Nineteen radiomic features were selected from each image type (FB or AIP) for analysis based on stability and variance. The selected FB and AIP radiomic feature sets had 6 common radiomic features between both image types and 13 unique features. The prognostic performances of the features for distant metastasis (DM) and locoregional recurrence (LRR) were evaluated using the concordance index (CI) and compared with two conventional features (tumor volume and maximum diameter). P-values were corrected for multiple testing using the false discovery rate procedure. None of the FB radiomic features were associated with DM, however, seven AIP radiomic features, that described tumor shape and heterogeneity, were (CI range: 0.638–0.676). Conventional features from FB images were not associated with DM, however, AIP conventional features were (CI range: 0.643–0.658). Radiomic and conventional multivariate models were compared between FB and AIP images using cross validation. The differences between the models were assessed using a permutation test. AIP radiomic multivariate models (median CI = 0.667) outperformed all other models (median CI range: 0.601–0.630) in predicting DM. None of the imaging features were prognostic of LRR. Therefore, image type impacts the performance of radiomic models in their association with disease recurrence. AIP images contained more information than FB images that were associated with disease recurrence in early stage NSCLC patients treated with SBRT, which suggests that AIP images may potentially be more optimal for the development of an imaging biomarker.


Frontiers in Oncology | 2016

Relationship between the Temporal Changes in Positron-Emission-Tomography-Imaging-Based Textural Features and Pathologic Response and Survival in Esophageal Cancer Patients

Stephen Yip; T Coroller; Nina N. Sanford; Harvey J. Mamon; Hugo J.W.L. Aerts; R Berbeco

Purpose Although change in standardized uptake value (SUV) measures and PET-based textural features during treatment have shown promise in tumor response prediction, it is unclear which quantitative measure is the most predictive. We compared the relationship between PET-based features and pathologic response and overall survival with the SUV measures in esophageal cancer. Methods Fifty-four esophageal cancer patients received PET/CT scans before and after chemoradiotherapy. Of these, 45 patients underwent surgery and were classified into complete, partial, and non-responders to the preoperative chemoradiation. SUVmax and SUVmean, two cooccurrence matrix (Entropy and Homogeneity), two run-length matrix (RLM) (high-gray-run emphasis and Short-run high-gray-run emphasis), and two size-zone matrix (high-gray-zone emphasis and short-zone high-gray emphasis) textures were computed. The relationship between the relative difference of each measure at different treatment time points and the pathologic response and overall survival was assessed using the area under the receiver-operating-characteristic curve (AUC) and Kaplan–Meier statistics, respectively. Results All Textures, except Homogeneity, were better related to pathologic response than SUVmax and SUVmean. Entropy was found to significantly distinguish non-responders from the complete (AUC = 0.79, p = 1.7 × 10−4) and partial (AUC = 0.71, p = 0.01) responders. Non-responders can also be significantly differentiated from partial and complete responders by the change in the run-length and size-zone matrix textures (AUC = 0.71–0.76, p ≤ 0.02). Homogeneity, SUVmax, and SUVmean failed to differentiate between any of the responders (AUC = 0.50–0.57, p ≥ 0.46). However, none of the measures were found to significantly distinguish between complete and partial responders with AUC <0.60 (p = 0.37). Median Entropy and RLM textures significantly discriminated patients with good and poor survival (log-rank p < 0.02), while all other textures and survival were poorly related (log-rank p > 0.25). Conclusion For the patients studied, temporal changes in Entropy and all RLM were better correlated with pathological response and survival than the SUV measures. The hypothesis that these metrics can be used as clinical predictors of better patient outcomes will be tested in a larger patient dataset in the future.


Physics in Medicine and Biology | 2016

Use of registration-based contour propagation in texture analysis for esophageal cancer pathologic response prediction

Stephen Yip; T Coroller; Nina N. Sanford; Elizabeth Huynh; Harvey J. Mamon; Hugo J.W.L. Aerts; R Berbeco

Change in PET-based textural features has shown promise in predicting cancer response to treatment. However, contouring tumour volumes on longitudinal scans is time-consuming. This study investigated the usefulness of contour propagation in texture analysis for the purpose of pathologic response prediction in esophageal cancer. Forty-five esophageal cancer patients underwent PET/CT scans before and after chemo-radiotherapy. Patients were classified into responders and non-responders after the surgery. Physician-defined tumour ROIs on pre-treatment PET were propagated onto the post-treatment PET using rigid and ten deformable registration algorithms. PET images were converted into 256 discrete values. Co-occurrence, run-length, and size zone matrix textures were computed within all ROIs. The relative difference of each texture at different treatment time-points was used to predict the pathologic responders. Their predictive value was assessed using the area under the receiver-operating-characteristic curve (AUC). Propagated ROIs from different algorithms were compared using Dice similarity index (DSI). Contours propagated by the fast-demons, fast-free-form and rigid algorithms did not fully capture the high FDG uptake regions of tumours. Fast-demons propagated ROIs had the least agreement with other contours (DSI = 58%). Moderate to substantial overlap were found in the ROIs propagated by all other algorithms (DSI = 69%-79%). Rigidly propagated ROIs with co-occurrence texture failed to significantly differentiate between responders and non-responders (AUC = 0.58, q-value = 0.33), while the differentiation was significant with other textures (AUC = 0.71-0.73, p < 0.009). Among the deformable algorithms, fast-demons (AUC = 0.68-0.70, q-value < 0.03) and fast-free-form (AUC = 0.69-0.74, q-value < 0.04) were the least predictive. ROIs propagated by all other deformable algorithms with any texture significantly predicted pathologic responders (AUC = 0.72-0.78, q-value < 0.01). Propagated ROIs using deformable registration for all textures can lead to accurate prediction of pathologic response, potentially expediting the temporal texture analysis process. However, fast-demons, fast-free-form, and rigid algorithms should be applied with care due to their inferior performance compared to other algorithms.

Collaboration


Dive into the T Coroller's collaboration.

Top Co-Authors

Avatar

Hugo J.W.L. Aerts

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Raymond H. Mak

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Vishesh Agrawal

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Ying Hou

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Stephanie W. Lee

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Huynh

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Vivek Narayan

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Stephen Yip

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

John Romano

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Chintan Parmar

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge