Network


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

Hotspot


Dive into the research topics where Patrick Grossmann is active.

Publication


Featured researches published by Patrick Grossmann.


Nature Communications | 2014

Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach.

Hugo J.W.L. Aerts; Emmanuel Rios Velazquez; R. Leijenaar; Chintan Parmar; Patrick Grossmann; S. Carvalho; Sara Cavalho; Johan Bussink; René Monshouwer; Benjamin Haibe-Kains; D. Rietveld; Frank Hoebers; C. René Leemans; Andre Dekker; John Quackenbush; Robert J. Gillies; Philippe Lambin

Human cancers exhibit strong phenotypic differences that can be visualized noninvasively by medical imaging. Radiomics refers to the comprehensive quantification of tumour phenotypes by applying a large number of quantitative image features. Here we present a radiomic analysis of 440 features quantifying tumour image intensity, shape and texture, which are extracted from computed tomography data of 1,019 patients with lung or head-and-neck cancer. We find that a large number of radiomic features have prognostic power in independent data sets of lung and head-and-neck cancer patients, many of which were not identified as significant before. Radiogenomics analysis reveals that a prognostic radiomic signature, capturing intratumour heterogeneity, is associated with underlying gene-expression patterns. These data suggest that radiomics identifies a general prognostic phenotype existing in both lung and head-and-neck cancer. This may have a clinical impact as imaging is routinely used in clinical practice, providing an unprecedented opportunity to improve decision-support in cancer treatment at low cost.


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.


Scientific Reports | 2015

Machine Learning methods for Quantitative Radiomic Biomarkers

Chintan Parmar; Patrick Grossmann; Johan Bussink; Philippe Lambin; Hugo J.W.L. Aerts

Radiomics extracts and mines large number of medical imaging features quantifying tumor phenotypic characteristics. Highly accurate and reliable machine-learning approaches can drive the success of radiomic applications in clinical care. In this radiomic study, fourteen feature selection methods and twelve classification methods were examined in terms of their performance and stability for predicting overall survival. A total of 440 radiomic features were extracted from pre-treatment computed tomography (CT) images of 464 lung cancer patients. To ensure the unbiased evaluation of different machine-learning methods, publicly available implementations along with reported parameter configurations were used. Furthermore, we used two independent radiomic cohorts for training (n = 310 patients) and validation (n = 154 patients). We identified that Wilcoxon test based feature selection method WLCX (stability = 0.84 ± 0.05, AUC = 0.65 ± 0.02) and a classification method random forest RF (RSD = 3.52%, AUC = 0.66 ± 0.03) had highest prognostic performance with high stability against data perturbation. Our variability analysis indicated that the choice of classification method is the most dominant source of performance variation (34.21% of total variance). Identification of optimal machine-learning methods for radiomic applications is a crucial step towards stable and clinically relevant radiomic biomarkers, providing a non-invasive way of quantifying and monitoring tumor-phenotypic characteristics in clinical practice.


Scientific Reports | 2015

Radiomic feature clusters and Prognostic Signatures specific for Lung and Head & Neck cancer

Chintan Parmar; R. Leijenaar; Patrick Grossmann; Emmanuel Rios Velazquez; Johan Bussink; D. Rietveld; Benjamin Haibe-Kains; Philippe Lambin; Hugo J.W.L. Aerts

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∓N) 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 & N cancer, respectively. These clusters were validated in independent external validation cohorts using rand statistic (Lung RS = 0.92, p < 0.001, H & N 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 & N CI = 0.68 ± 0.01; Lung histology AUC = 0.56 ± 0.03, Lung stage AUC = 0.61 ± 0.01, H & N HPV AUC = 0.58 ± 0.03, H & N 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.


Frontiers in Oncology | 2015

radiomic Machine-learning classifiers for Prognostic Biomarkers of head and neck cancer

Chintan Parmar; Patrick Grossmann; D. Rietveld; Philippe Lambin; Hugo J.W.L. Aerts

Introduction “Radiomics” extracts and mines a large number of medical imaging features in a non-invasive and cost-effective way. The underlying assumption of radiomics is that these imaging features quantify phenotypic characteristics of an entire tumor. In order to enhance applicability of radiomics in clinical oncology, highly accurate and reliable machine-learning approaches are required. In this radiomic study, 13 feature selection methods and 11 machine-learning classification methods were evaluated in terms of their performance and stability for predicting overall survival in head and neck cancer patients. Methods Two independent head and neck cancer cohorts were investigated. Training cohort HN1 consisted of 101 head and neck cancer patients. Cohort HN2 (n = 95) was used for validation. A total of 440 radiomic features were extracted from the segmented tumor regions in CT images. Feature selection and classification methods were compared using an unbiased evaluation framework. Results We observed that the three feature selection methods minimum redundancy maximum relevance (AUC = 0.69, Stability = 0.66), mutual information feature selection (AUC = 0.66, Stability = 0.69), and conditional infomax feature extraction (AUC = 0.68, Stability = 0.7) had high prognostic performance and stability. The three classifiers BY (AUC = 0.67, RSD = 11.28), RF (AUC = 0.61, RSD = 7.36), and NN (AUC = 0.62, RSD = 10.52) also showed high prognostic performance and stability. Analysis investigating performance variability indicated that the choice of classification method is the major factor driving the performance variation (29.02% of total variance). Conclusion Our study identified prognostic and reliable machine-learning methods for the prediction of overall survival of head and neck cancer patients. Identification of optimal machine-learning methods for radiomics-based prognostic analyses could broaden the scope of radiomics in precision oncology and cancer care.


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.


Frontiers in Oncology | 2016

Exploratory Study to Identify Radiomics Classifiers for Lung Cancer Histology

Weimiao Wu; Chintan Parmar; Patrick Grossmann; John Quackenbush; Philippe Lambin; Johan Bussink; Raymond H. Mak; Hugo J.W.L. Aerts

Background Radiomics can quantify tumor phenotypic characteristics non-invasively by applying feature algorithms to medical imaging data. In this study of lung cancer patients, we investigated the association between radiomic features and the tumor histologic subtypes (adenocarcinoma and squamous cell carcinoma). Furthermore, in order to predict histologic subtypes, we employed machine-learning methods and independently evaluated their prediction performance. Methods Two independent radiomic cohorts with a combined size of 350 patients were included in our analysis. A total of 440 radiomic features were extracted from the segmented tumor volumes of pretreatment CT images. These radiomic features quantify tumor phenotypic characteristics on medical images using tumor shape and size, intensity statistics, and texture. Univariate analysis was performed to assess each feature’s association with the histological subtypes. In our multivariate analysis, we investigated 24 feature selection methods and 3 classification methods for histology prediction. Multivariate models were trained on the training cohort and their performance was evaluated on the independent validation cohort using the area under ROC curve (AUC). Histology was determined from surgical specimen. Results In our univariate analysis, we observed that fifty-three radiomic features were significantly associated with tumor histology. In multivariate analysis, feature selection methods ReliefF and its variants showed higher prediction accuracy as compared to other methods. We found that Naive Baye’s classifier outperforms other classifiers and achieved the highest AUC (0.72; p-value = 2.3 × 10−7) with five features: Stats_min, Wavelet_HLL_rlgl_lowGrayLevelRunEmphasis, Wavelet_HHL_stats_median, Wavelet_HLL_stats_skewness, and Wavelet_HLH_glcm_clusShade. Conclusion Histological subtypes can influence the choice of a treatment/therapy for lung cancer patients. We observed that radiomic features show significant association with the lung tumor histology. Moreover, radiomics-based multivariate classifiers were independently validated for the prediction of histological subtypes. Despite achieving lower than optimal prediction accuracy (AUC 0.72), our analysis highlights the impressive potential of non-invasive and cost-effective radiomics for precision medicine. Further research in this direction could lead us to optimal performance and therefore to clinical applicability, which could enhance the efficiency and efficacy of cancer care.


Neuroradiology | 2015

Somatic mutations associated with MRI-derived volumetric features in glioblastoma

David A. Gutman; William D. Dunn; Patrick Grossmann; Lee A. D. Cooper; Chad A. Holder; Keith L. Ligon; Brian M. Alexander; Hugo J.W.L. Aerts

IntroductionMR imaging can noninvasively visualize tumor phenotype characteristics at the macroscopic level. Here, we investigated whether somatic mutations are associated with and can be predicted by MRI-derived tumor imaging features of glioblastoma (GBM).MethodsSeventy-six GBM patients were identified from The Cancer Imaging Archive for whom preoperative T1-contrast (T1C) and T2-FLAIR MR images were available. For each tumor, a set of volumetric imaging features and their ratios were measured, including necrosis, contrast enhancing, and edema volumes. Imaging genomics analysis assessed the association of these features with mutation status of nine genes frequently altered in adult GBM. Finally, area under the curve (AUC) analysis was conducted to evaluate the predictive performance of imaging features for mutational status.ResultsOur results demonstrate that MR imaging features are strongly associated with mutation status. For example, TP53-mutated tumors had significantly smaller contrast enhancing and necrosis volumes (p = 0.012 and 0.017, respectively) and RB1-mutated tumors had significantly smaller edema volumes (p = 0.015) compared to wild-type tumors. MRI volumetric features were also found to significantly predict mutational status. For example, AUC analysis results indicated that TP53, RB1, NF1, EGFR, and PDGFRA mutations could each be significantly predicted by at least one imaging feature.ConclusionMRI-derived volumetric features are significantly associated with and predictive of several cancer-relevant, drug-targetable DNA mutations in glioblastoma. These results may shed insight into unique growth characteristics of individual tumors at the macroscopic level resulting from molecular events as well as increase the use of noninvasive imaging in personalized medicine.


Nucleic Acids Research | 2016

Integration of TP53, DREAM, MMB-FOXM1 and RB-E2F target gene analyses identifies cell cycle gene regulatory networks

Martin Fischer; Patrick Grossmann; Megha Padi; James A. DeCaprio

Cell cycle (CC) and TP53 regulatory networks are frequently deregulated in cancer. While numerous genome-wide studies of TP53 and CC-regulated genes have been performed, significant variation between studies has made it difficult to assess regulation of any given gene of interest. To overcome the limitation of individual studies, we developed a meta-analysis approach to identify high confidence target genes that reflect their frequency of identification in independent datasets. Gene regulatory networks were generated by comparing differential expression of TP53 and CC-regulated genes with chromatin immunoprecipitation studies for TP53, RB1, E2F, DREAM, B-MYB, FOXM1 and MuvB. RNA-seq data from p21-null cells revealed that gene downregulation by TP53 generally requires p21 (CDKN1A). Genes downregulated by TP53 were also identified as CC genes bound by the DREAM complex. The transcription factors RB, E2F1 and E2F7 bind to a subset of DREAM target genes that function in G1/S of the CC while B-MYB, FOXM1 and MuvB control G2/M gene expression. Our approach yields high confidence ranked target gene maps for TP53, DREAM, MMB-FOXM1 and RB-E2F and enables prediction and distinction of CC regulation. A web-based atlas at www.targetgenereg.org enables assessing the regulation of any human gene of interest.


Scientific Reports | 2016

Defining a Radiomic Response Phenotype: A Pilot Study using targeted therapy in NSCLC.

Hugo J.W.L. Aerts; Patrick Grossmann; Yongqiang Tan; Geoffrey R. Oxnard; Naiyer A. Rizvi; Lawrence H. Schwartz; Binsheng Zhao

Medical imaging plays a fundamental role in oncology and drug development, by providing a non-invasive method to visualize tumor phenotype. Radiomics can quantify this phenotype comprehensively by applying image-characterization algorithms, and may provide important information beyond tumor size or burden. In this study, we investigated if radiomics can identify a gefitinib response-phenotype, studying high-resolution computed-tomography (CT) imaging of forty-seven patients with early-stage non-small cell lung cancer before and after three weeks of therapy. On the baseline-scan, radiomic-feature Laws-Energy was significantly predictive for EGFR-mutation status (AUC = 0.67, p = 0.03), while volume (AUC = 0.59, p = 0.27) and diameter (AUC = 0.56, p = 0.46) were not. Although no features were predictive on the post-treatment scan (p > 0.08), the change in features between the two scans was strongly predictive (significant feature AUC-range = 0.74–0.91). A technical validation revealed that the associated features were also highly stable for test-retest (mean ± std: ICC = 0.96 ± 0.06). This pilot study shows that radiomic data before treatment is able to predict mutation status and associated gefitinib response non-invasively, demonstrating the potential of radiomics-based phenotyping to improve the stratification and response assessment between tyrosine kinase inhibitors (TKIs) sensitive and resistant patient populations.

Collaboration


Dive into the Patrick Grossmann's collaboration.

Top Co-Authors

Avatar

Hugo J.W.L. Aerts

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Philippe Lambin

Maastricht University Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Chintan Parmar

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Benjamin Haibe-Kains

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

R. Leijenaar

Maastricht University Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Raymond H. Mak

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
Top Co-Authors

Avatar

Johan Bussink

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Robert J. Gillies

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge