Published OnlineFirst July 27, 2018; DOI: 10.1158/1078-0432.CCR-17-3420

Precision Medicine and Imaging Clinical Cancer Research A Coclinical Radiogenomic Validation Study: Conserved Magnetic Resonance Radiomic Appearance of Periostin-Expressing Glioblastoma in Patients and Xenograft Models Pascal O. Zinn1,2,3,4, Sanjay K. Singh2,5, Aikaterini Kotrotsou5, Islam Hassan5, Ginu Thomas5, Markus M. Luedi2,6, Ahmed Elakkad5, Nabil Elshafeey5, Tagwa Idris5, Jennifer Mosley2, Joy Gumin3, Gregory N. Fuller7, John F. de Groot8, Veera Baladandayuthapani9, Erik P. Sulman10, Ashok J. Kumar5, Raymond Sawaya3, Frederick F. Lang3, David Piwnica-Worms2, and Rivka R. Colen2,5

Abstract

Purpose: Radiomics is the extraction of multidimensional Results: Our robust pipeline consists of segmentation, imaging features, which when correlated with genomics, is radiomic-feature extraction, feature normalization/selection, termed radiogenomics. However, radiogenomic biological and predictive modeling. The combination of skull stripping, validation is not sufficiently described in the literature. brain-tissue focused normalization, and patient-specific nor- We seek to establish causality between differential malization are unique to this study, providing comparable expression status and MRI-extracted radiomic-features in cross-platform, cross-institution radiomic features. POSTN glioblastoma. expression status was not associated with qualitative or vol- Experimental Design: Radiogenomic predictions and val- umetric MRI parameters. Radiomic features significantly pre- idation were done using the Cancer Genome Atlas and Repos- dicted POSTN expression status in patients (AUC: 76.56%; itory of Molecular Brain Neoplasia Data glioblastoma patients sensitivity/specificity: 73.91/78.26%) and OX (AUC: 92.26%; (n ¼ 93) and orthotopic xenografts (OX; n ¼ 40). Tumor sensitivity/specificity: 92.86%/91.67%). Furthermore, radio- phenotypes were segmented, and radiomic-features extracted mic features in OX were significantly associated with patients using the developed radiome-sequencing pipeline. Patients with similar POSTN expression levels (AUC: 93.36%; sensi- and animals were dichotomized on the basis of Periostin tivity/specificity: 82.61%/95.74%; P ¼ 02.021E15). (POSTN) expression levels. RNA and levels confirm- Conclusions: We determined causality between radiomic ed RNAi-mediated POSTN knockdown in OX. Total RNA of texture features and POSTN expression levels in a preclinical tumor cells isolated from mouse brains (knockdown and model with clinical validation. Our biologically validated control) was used for microarray-based expression profiling. radiomic pipeline also showed the potential application Radiomic-features were utilized to predict POSTN expression for human–mouse matched coclinical trials. Clin Cancer Res; status in patient, mouse, and interspecies. 24(24); 6288–99. 2018 AACR.

Introduction post-care follow-up. However, despite recent exponential refine- Imaging has transformed the medical field by providing a ments in imaging technologies in terms of acquisition time and noninvasive method to interrogate the human body and under- resolution, we have barely begun to tap the potential of imaging to lying biological processes. Particularly in patients with cancer, characterize tissues or tumors beyond qualitative description or imaging plays a key role throughout the entire treatment para- gross tumor size on routine imaging sequences. In fact, all current digm ranging from diagnosis and assessing treatment response to imaging assessment criteria [such as RECIST, response assessment

1Department of Neurosurgery, Baylor College of Medicine, Houston Texas. Houston, Texas. 10Department of Radiation Oncology, The University of Texas 2Department of Cancer Systems Imaging, The University of Texas MD Anderson MD Anderson Cancer Center, Houston, Texas. Cancer Center, Houston, Texas. 3Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas. 4Department of Cancer Note: Supplementary data for this article are available at Clinical Cancer Biology, Division of Basic Science Research, The University of Texas MD Research Online (http://clincancerres.aacrjournals.org/). Anderson Cancer Center, Houston, Texas. 5Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston Texas. 6Depart- Corresponding Author: Rivka R. Colen, The University of Texas MD Anderson ment of Anesthesiology, Bern University Hospital Inselspital, University of Bern, Cancer Center, 3SCRB4.3606, Unit 1907, 1881 East Road, Houston, TX 77054. Bern, Switzerland. 7Department of Pathology, Section Neuropathology, The Phone: 713-745-8552; Fax: 713-794-5456; E-mail: [email protected] University of Texas MD Anderson Cancer Center, Houston, Texas. 8Department of Neuro-Oncology, Division of Cancer Medicine, The University of Texas MD doi: 10.1158/1078-0432.CCR-17-3420 Anderson Cancer Center, Houston, Texas. 9Department of Biostatistics, Division of Quantitative Sciences, The University of Texas MD Anderson Cancer Center, 2018 American Association for Cancer Research.

6288 Clin Cancer Res; 24(24) December 15, 2018

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 27, 2018; DOI: 10.1158/1078-0432.CCR-17-3420

Validation of Radiomics and Radiogenomics

(8, 9). Recent studies have demonstrated an association between Translational Relevance imaging features (also termed phenotypes) and cancer histology, Radiomics, the automated high-throughput extraction of tumor grades, and genomics (5, 10–13). The linkage of imaging multidimensional imaging features, captures microscale infor- phenotypes with genomic data is termed radiogenomics (also mation hidden within conventional imaging beyond what is termed imaging genomics; ref. 10). Fundamental to radioge- visible to the naked human eye. The linkage of imaging nomics is the hypothesis that expression of specific sets of phenotypes with genomic data is termed radiogenomics. or driver mutations impact the extractable imaging features, but Fundamental to radiogenomics is the hypothesis that expres- this has not been directly demonstrated in a highly robust model. sion of specific sets of genes or driver mutations impacts the Imaging-molecular connections must be established in a extractable imaging features, but this has not been directly robust manner, specifically by establishing causality (as opposed demonstrated in a highly robust model; to date, the linkages to correlations), for its acceptance as a validated tool. To establish between imaging and genomics remain at a correlative stage causality, a closed system is required for in vivo manipulation of and lack established causality. In this study, we seek to address genomic expression patterns and linkage to radiomic features this gap in knowledge; we bring forward an approach for using a preclinical model system. A few studies have tried to integrated end-to-end methodology for extraction of imaging elucidate the biological significance of imaging characteristics radiomic features and validation in both a preclinical closed (14, 15). Joo and colleagues demonstrated that xenograft tumors model system and patient cohort. Further, we demonstrate produced from patient-derived glioblastoma (GBM) cell lines the potential of radiomics in coclinical trials. To our knowl- were similar to their paired human parental tumors (14). In the edge, this is the first study functionally validating imaging- latter study, they found the invasiveness of the parental tumor on molecular features. MRI correlated with invasive measures based on paraffin histo- pathology sections of the xenograft tumor. In a different study using a heterotopic colorectal cancer mouse model, Panth and colleagues found changes in CT imaging features after induced alterations in gene expression and radiation treatment (15). in neuro-oncology (RANO), immune related RECIST (irRECIST), However, to date, the linkages between imaging and genomics and immune related response criteria (irRC)] used to evaluate largely remain at a correlative stage and lack established causality tumor response in the clinical setting and in clinical trials are in a model system relevant for human disease. dependent on changes in tumor size and do not accurately capture In this study, we seek to address this gap in knowledge, which responses to therapy (1–4). With the development of immune can be expected to change patient management. To our knowl- and molecularly targeted therapies, alternative imaging assess- edge, this is the first study to establish causality between imaging- ment criteria and markers are needed that go beyond mere molecular (radiogenomic) features. In this study, we bring for- changes in tumor size. In fact, with some therapies such as ward an approach for an integrated end-to-end methodology immunotherapy, positive response to treatment is associated with for extraction of imaging radiomic features and its validation in initial decrease in tumor size in only 10% of patients, whereas both a preclinical closed model system and in a clinical patient other positive responders fail to show such a measurable initial cohort. We use the most common primary malignant brain decline in tumor size (4). This uncoupling of tumor metrics and tumor characterized by extreme heterogeneity and poor patient response to therapy is a clinical dilemma and challenge for survival (16–18), GBM, as the solid cancer prototype in this clinicians. Furthermore, the specific "makeup" of a tumor's imag- study. Periostin (POSTN) was used as the gene prototype, given ing characteristics reflects underlying molecular processes, which its previous radiogenomic description in the first published cannot be evaluated using changes in tumor size alone. Unlocking quantitative radiogenomic study in glioma of The Cancer the full breadth of data contained in routine imaging studies Genome Atlas (TCGA; ref. 13); its more recent surfacing as an would enable the identification of high quality, noninvasive, important gene involved in GBM invasion (19), one of the most clinically relevant, and actionable imaging markers. important causes of GBM recurrence; and its potential use as a Radiomics, an emerging field within imaging, is poised to do GBM therapeutic target in developing clinical trials (20). just that. An automated high-throughput extraction of multidi- mensional imaging features (on a pixel and voxel level), radio- mics captures microscale information hidden within convention- Materials and Methods al imaging and beyond what is visible to the naked human eye Patient-derived imaging, clinical and genomic material (5–7). Like the layer of complexity and resolution that genomics were collected from TCGA, The Cancer Imaging Archive has added to tumor biology, radiomics similarly adds to conven- (TCIA), and the Repository of Molecular Brain Neoplasia Data tional imaging. However, a critical need exists to understand the (REMBRANDT); all patient data collection were HIPAA compliant biological underpinnings of imaging, specifically radiomics, for and approved by MD Anderson Cancer Center's (MDACC, Hous- informed clinical decision making. This also necessitates a proper ton, TX) Institutional Review Board (IRB; PA130720). Analyses understanding of how changes in genomics affect imaging infor- were based on a total of 79 TCGA patients and 14 REMBRANDT mation. With radiomics, it is now realistic to begin testing precise patients for whom complete baseline MRI data and POSTN gene linkages and investigate causality between imaging and genomics. expression data were available. Detailed demographic informa- Understanding the molecular bedrocks of radiomics is important tion is presented in Tables 1 and 2. due to imaging's inherent advantages; in contrast to genomic For the preclinical portion of the study, MD Anderson Cancer analyses which are often performed on only a fraction of the Center (MDACC) patient-derived glioma stem cells (GSC) lines tumor, an advantage of imaging is that it assesses the entire three- were isolated in accordance with a protocol (LAB04-0001) dimensional tumor volume inclusive of spatial heterogeneity approved by MDACC's IRB. Patient consent was obtained. For

www.aacrjournals.org Clin Cancer Res; 24(24) December 15, 2018 6289

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 27, 2018; DOI: 10.1158/1078-0432.CCR-17-3420

Zinn et al.

Table 1. TCGA patient demographic table Patients with high POSTN level Patients with low POSTN level expression (n ¼ 39) expression (n ¼ 40) Age (years) 60 24 63 18 Sex (male/female) 28 (71.7%)/11 (28.2%) 23 (57.5%)/17 (42.5%) KPS (80/<80/not reported) 25 (64.1%)/4 (10.2%)/9 (23) 29 (72.5%)/6 (15%)/5 (12.5%)

the cell study, the IRB issued a waiver of informed consent since contrast-enhanced T1WI into the same geometric space. Images data collection was retrospective. All animal experiments com- were registered using the General Registrations (BRAINS) Tool- plied with institutional regulations and were approved by box in 3D Slicer (affine registration, 12 degrees of freedom; MDACC's Institutional Animal Care and Use Committee (proto- Interpolation mode: Nearest Neighbor; https://www.slicer.org/ col 00001100) in accordance with the guidelines of the American wiki/Documentation/4.8/Registration/RegistrationLibrary), and Association for Laboratory Animal Science. the transformation matrix that maps each point of the reference image to the target's image space was obtained. An adequate Image segmentation registration was designated as having an error of 2 mm or less; all Image analysis and software. We used 3D Slicer version 4.3.1 patient data were reviewed post-registration and if the error was (www.slicer.org), an open-source image analytics platform, for more than 2 mm then the patient was not included in the study. tumor segmentation (21–23). The segmented images were After image registration, images were segmented from the reviewed in consensus by two board-certified neuroradiologists periphery (edema/invasion) to center (necrosis). Three distinct with 9 (R.R. Colen) and 35 (A.J. Kumar) years of experience. For imaging phenotypes were segmented semiautomatically: edema/ the segmentation of the mouse MRI, we followed an approach invasion, active enhancing tumor, and necrosis. A region of the similar to that used for the patient imaging analysis using 3D contralateral normal-appearing white matter (opposite hemi- Slicer software. In brief, conventional MRI series were coregistered sphere and opposite lobe with respect to a line perpendicular to bring all mouse images to same space, using a registration to the midline) was also segmented for within-sequence nor- method identical to the method used in humans (detailed malization of the data. Hemorrhage appearing as hyperintensity below). Then, we outlined different tumor phenotypes including on precontrast T1WI and contrast-enhancing vessels were sub- edema, enhancement, and necrosis on the corresponding imaging tracted from the FLAIR region to prevent contamination of the series similar to the approach used in humans (detailed below). radiomic features obtained thereafter. The outlines of all seg- The image analysis team was blinded to the patient cohort and mented phenotypes were saved in a label volume. Finally, the animal groups (control vs. knockdown). inverted transformation matrix was applied to the label volume using nearest-neighbor interpolation to propagate the label Volume selection. We used contrast-enhanced axial T1-weighted volume to the original image. The volume of each phenotype imaging (T1WI) sequences and precontrast axial Fluid-Attenuated was calculated by multiplying the number of voxels in a phe- Inversion Recovery (FLAIR) sequences. Contrast-enhanced T1WI notype with the voxel volume. was used for segmentation of the enhancing component (i.e., active enhancing tumor) and nonenhancing central tumor com- Radiome sequencing textural analysis ponent (i.e., necrosis). The edema/invasion portion was segment- Image preprocessing. An overview of the approach used for radio- ed using the FLAIR sequence; this region was assessed based on the mic feature extraction is shown in Fig. 1B. All MRI scans were peritumoral hyperintensity seen on the FLAIR sequence (Fig. 1A). processed using the same pipeline. In the first step, we used Hyperintensity on precontrast T1WI was used to assess hemor- FMRIB's Brain Extraction Tool (BET; http://fsl.fmrib.ox.ac.uk/ rhage. Of the 93 TCGA/TCIA/REMBRANDT patients, 1 was fsl/fslwiki/BET) to remove nonbrain tissue from the anatomical scanned with a 1T system (1.08%), 57 were scanned with a MR images (Fig. 1B; refs. 25, 26). Contrast-enhanced T1WI was 1.5T system (61.3%), and 26 were scanned with a 3T system used as an input to BET. In all runs, the BET option for "bias field (28%). Scanner strength information was not available for 9 correction and neck removal" was selected. The resulting brain patients (9.7%). Standard imaging parameters (slice thickness, mask from BET was applied to the FLAIR sequences, thus remov- voxel size, and slice gap) were used for each sequence (Supple- ing nonbrain tissue from those images also. In the second step, to mentary Table S1A and S1B). account for scanner differences, we applied the Nyul intensity normalization algorithm to standardize the intensity scales Image registration, segmentation, and model making. Image reg- across MR images of the same contrast (brain-specific istration and segmentation were done as previously published by normalization; Fig. 1B; refs. 27–29). In brief, Nyul normalization our group (24). Prior to image segmentation, within-patient involves the matching of the histograms between a reference image registration was performed to align the FLAIR imaging and image and the image of each subject in the cohort. In our

Table 2. Rembrandt patient demographic tablea Patients with high POSTN Patients with low POSTN level expression (n ¼ 7) level expression (n ¼ 7) Age (years) 59 12 51.5 5 Sex (male/female/not reported) 3 (43%)/2 (28.5%)/2 (28.5) 1 (14.25%)/1 (14.25)/5 (71.5%) 0.552 KPS (80/not reported) 4 (57%)/3 (43%) 2 (28.5%)/5 (71.5%) 0.473 aDemographic information was missing for 7 of the 14 patients used from Rembrandt database.

6290 Clin Cancer Res; 24(24) December 15, 2018 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 27, 2018; DOI: 10.1158/1078-0432.CCR-17-3420

Validation of Radiomics and Radiogenomics

Figure 1. Radiome sequencing pipeline for solid tumor (glioblastoma) and tissue characterization. A, Semiautomated segmentation of the three imaging phenotypes: necrosis (left), postcontrast active enhancing tumor (middle), and fluid attenuated inversion recovery (FLAIR) peritumoral edema/invasion, representing edematous tumor and sites of cellular invasion into brain tissue (right). B, Automated segmentation-based radiomic feature extraction is followed by patient-specific normalization and feature selection, which consists of the normalization of contralateral normal-appearing white matter as an internal control normalizer to account for various potential intra- and interinstitutional biases, a crucial step that ensures comparability. The next step is volume-dependent feature generation, which uses the necrosis, postcontrast enhancement, and FLAIR volumes for the corresponding radiomic feature sets acquired from the respective sequences, thus doubling the amount of radiomic features and creating a set of tumor volume–independent and –dependent radiomic features. Finally, this homogenous dataset can then enter feature selection and predictive modeling for any GBM trait of interest.

implementation, firstly, the MR images (FLAIR and contrast- level histogram is a function showing the number of voxels that enhanced T1WI) of one patient from the cohort were randomly have a specific intensity in the original volume of interest; thus, the selected as the reference, and the corresponding histograms extracted features provide information about the intensity distri- were obtained; the intensities of the tumor were excluded from bution within the volume of interest. From each intensity-level the calculation of the histogram, because the tumor is an histogram, the following 10 features were extracted: minimum, abnormal environment. In the next step, for each patient in maximum, mean, SD, skewness, kurtosis, and four percentiles the cohort, the histograms were mapped via a piecewise linear (1%, 5%, 95%, 99%; refs. 31, 32). transformation to the reference histogram; thus, the resulting GLCM is a matrix computed from the original volume of transformed image has similar range of intensities. Finally, we interest, and each element of the matrix Ai,j represents the joint extracted radiomic textural features from each phenotype (i.e., probability of two pixels with gray levels i and j with distance d edema/invasion, active enhancing tumor, and necrosis, and apart and a certain angular direction u. In our implementation, the contralateral normal-appearing white matter) as well as the GLCMs were calculated in four angular directions corresponding whole tumor volume on the FLAIR sequences and contrast- to in-plane rotations (u ¼ 0,45,90, 135) and d ¼ 1 voxel (Fig. enhanced T1WI. We followed an identical approach for proces- 1B). Each GLCM is normalized to the sum of co-occurrence pairs; sing the mouse MRI prior to radiomic feature calculation, thus, the extracted features are independent of the number of except that we omitted the skull-stripping step. original observations. From each GLCM, the following 20 texture features were extracted: autocorrelation, contrast, correlation, Textural radiome sequencing. Whole radiome sequencing was cluster shade, cluster prominence, dissimilarity, energy, entropy, performed, and the radiomic features were extracted for each homogeneity, maximum probability, variance, sum average, sum phenotype and whole-tumor volume, using: (i) intensity-level variance, sum entropy, difference variance, difference entropy, histogram (first-order features), and (ii) gray-level co-occurrence information measure of correlation 1 and 2, inverse difference matrices (GLCMs; second-order features; ref. 30). The intensity- moment, and normalized inverse difference moment (30, 33, 34).

www.aacrjournals.org Clin Cancer Res; 24(24) December 15, 2018 6291

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 27, 2018; DOI: 10.1158/1078-0432.CCR-17-3420

Zinn et al.

To obtain invariant measures of the features across different Radiomic feature selection. Given the large number of features rotations, we calculated the average, range, and angular variance provided by radiomics, feature selection is the first step in the of each feature for different u, thereby resulting in 60 rotation- pipeline. For feature selection, we used Least Absolute Shrinkage invariant texture features for each volume of interest for every gray and Selection Operator (LASSO) regularization (L1 regulariza- level. tion) technique (35). LASSO is a regression analysis method that In our implementation, the original images were discretized typically performs variable selection and regularization to into N number of gray levels, where n ¼ 8, 16, 32, 64, or 256. enhance the prediction accuracy and avoid overfitting of the Reducing the number of gray levels increases the signal-to-noise features on the model. L1 regularization coupled with early ratio and eliminates sparseness in the final GLCM. Intensity-level stopping (at one third of the patient size) was used for feature histogram-based features were calculated using images discretized selection to select the most important and relevant features at 256 gray levels and GLCM-based features were calculated using required for the model building. Later, these important features all gray levels. In summary, our analysis resulted in a total of 310 were used for model building. We used the latter models in the radiomic features for each phenotype per MRI sequence. Taking prediction of high versus low POSTN mice and human. The into account the two MRI sequences (contrast-enhanced T1WI criteria of groupings into high versus low POSTN gene expression and T2/FLAIR) along with the phenotypes (edema/invasion, levels were based on cohort (n ¼ 93) specific median expression enhancing active tumor, and necrosis) and whole-tumor volume, levels. a total of 2,480 radiomic features (2,400 second-order and 80 first The reliability of our radiomic pipeline was assessed by intra- order features) were extracted from human GBM. An additional class correlation coefficient (ICC) values across institutions, scan- 620 radiomic features from the contralateral normal-appearing ner, etc., before and after normalization steps for the features in white matter were extracted from human MRIs and used for the final predictive model. In addition, the nonnormalized fea- within-sequence normalization purposes. Similarly, in the case tures were tested for their ability to predict the outcome. of mice, a total of 2,480 (2,400 second-order and 80 first-order features) features were obtained. Patient and mice multivariable predictive modeling. Radiomic feature selection and classification model building was performed Extracted feature generation. Of the 310 radiomic features, a total by implementing gradient boosting from the XGboost package of 300 MRI radiomic features per phenotype (enhancing active (eXtreme Gradient Boosting) and works by implementing gradi- tumor, edema/invasion, necrosis, and whole GBM tumor) ent boosting. The XGboost algorithm works by converting weak extracted per each imaging sequence (T1 and T2/FLAIR) were learners to strong learners using the boosting technique, in which GLCM-based features as described above. This represents a total of trees are developed with the help of information from the pre- 2,400 GLCM-based features extracted per tumor per patient (as viously grown trees and passing that information from one tree to well as per animal). To consider tumor volumes, 2,400 new another iteratively. Hence, slowly the trees to learn from the data variables (xij) were added by dividing each of these feature (f'ij) and improve its prediction by minimizing the loss function or by the corresponding phenotype volume. As a result, 4,880 mis-classification error rate. XGboost enables parallel computa- (volume-dependent and volume-independent) features (2,400 tion, regularization, cross validation, missing value imputation original second-order, 2,400 volume-dependent second-order (if needed), and tree pruning (36). and 80 first-order features) per tumor per patient (animal) were For classification and model building, we used gbtree booster obtained. of XGboost and built binary logistic regression model to classify patients into high and low POSTN groups based on the selected Contralateral normalization. To account for differences in the radiomic features resulted from LASSO regularization. We per- image acquisition hardware and software used across TCGA formed hyperparameter tuning required for tree booster and institutions, we performed a patient-specific contralateral hemi- cross-validation using leave one out cross-validation (LOOCV) sphere normalization of the features using the features extracted over 100 iterations (n rounds ¼ 100) to avoid overfitting and from the contralateral normal-appearing white matter. Conse- acquire best prediction model that has better accuracy, AUC, quently, each feature (fij) was subtracted from the corresponding sensitivity, specificity and P values. contralateral radiomic feature fij(wm). We assessed the perfor- Besides LOOCV, a 50% random split was used to train and test mance of the patient-specific contralateral hemisphere normali- the model for outcome. ROC analysis and analysis with a 95% zation of the features across institutions, MRI scanner brands, and confidence interval (CI) were implemented to measure the model magnetic field strengths by plotting pre- and post- normalization accuracy. AUC, sensitivity, specificity, PPV, NPV, and P values for feature distribution. each LOOCV and prediction output were calculated.

Biostatistical radiomic analysis: feature selection, Translational causality investigations classification, and predictive modeling Two independent GSC lines upon validation of efficient short We used R software (version 3.4.0, R Foundation for Statistical hairpin RNA (shRNA)-mediated POSTN knockdown were used to Computing, Vienna, Austria) for all the statistical analysis: develop orthotopic tumors in mouse brains. See Supplementary package xgboost (version 0.6.4.1) for the feature selection task Materials and Methods for details. In brief, leveraging a controlled and the Machine Learning package mlr (version 2.11) to build preclinical experimental system for mechanistic validation of the binary logistic model of XGboost using "gbtree" booster for imaging genomics, we illustrate the potential for radiogenomic classification. Finally, ROC analysis was performed using pROC causality beyond mere correlation. First, we screened 47 GSC lines package (version 1.9.1). AUC, sensitivity, specificity, PPV, NPV that met three criteria: high POSTN expression, expression of and P values are reported for each LOOCV and prediction stemness markers, and their ability to produce stable orthotopic outputs. xenograft tumor using primary GSC lines to most faithfully

6292 Clin Cancer Res; 24(24) December 15, 2018 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 27, 2018; DOI: 10.1158/1078-0432.CCR-17-3420

Validation of Radiomics and Radiogenomics

replicate human GBM (Supplementary Fig. S1). First, we used comparable cross-platform, cross-institution radiomic features gene expression microarray and quantitative reverse transcriptase as demonstrated qualitatively in TCGA pre- and post- patient- PCR (qRT-PCR) to select two GSC lines (GSC11 and GSC126) specific normalization plots (Supplementary Figs. S4–S6). This which met the latter criteria. We then transduced these GSC lines novel pipeline was used for all imaging and statistical radiomic with lentiviruses containing a doxycycline-inducible shRNA and radiogenomic analysis in our study. against POSTN, which coexpressed GFP via an internal ribosome entry site (IRES), thereby allowing visualization/isolation of Qualitative and quantitative (volumetric) MRI is unable to knockdown cells both in vitro and ex vivo (Supplementary classify GBM based on prototype POSTN gene expression level Fig. S2) and selected puromycin resistant stable clones with in patients and animal models significant knockdown of POSTN at both the messenger RNA We evaluated POSTN expression levels in mesenchymal and protein levels (Supplementary Fig. S3). These clones were (MES) and proneural (PN) subgroups of GBM and found that orthotopically implanted into nude mice (n ¼ 40). MRI was used expression of POSTN is highly correlated with MES subgroup to confirm the growth of the xenograft tumors (Fig. 3E). Ex vivo (Fig. 2A). Gene set enrichment analysis (GSEA) of whole þ flow-sorted cells from the control (APC ve) and POSTN-knock- genome expression profiles of patient with high POSTN vs. þ þ down (APC veGFP ve) mouse tumors (Supplementary Fig. S2) low POSTN expression (grouped according to median cutoff), were subjected to RNA extraction and qRT-PCR for POSTN. Those revealed that high POSTN expression was significantly corre- cells from knockdown tumors showed a significant decrease in lated with hallmarks such as epithelial mesenchymal transition POSTN mRNA levels (Fig. 3A); we also performed xenograft (p-value < 0.001, FDR q-value: 0), angiogenesis (p-value < tumor IHC to confirm that the POSTN-knockdown tumors had 0.001, FDR q-value: 0.001) etc. (Fig. 2B). As suggested by significantly lower POSTN protein expression than that of control GSEA, patients with high endothelial proliferation, as extracted tumors (Supplementary Fig. S3). from Cancer digital slide archive (http://cancer.digitalslidearc hive.net/; n ¼ 232; ref. 43) displayed significantly higher levels of POSTN expression (P ¼ 0.0027) in comparison to that with Results low endothelial proliferation (n ¼ 152; Fig. 2C and D). How- Robust high-throughput radiomic and radiogenomic pipeline ever, qualitative and quantitative volumetric analyses of MRI We established a robust image analytical pipeline consisting of scans of patients with high and low POSTN expression image segmentation, radiomic feature extraction, feature normal- appeared indistinguishable (Fig. 2E) and illustrated statistically ization and selection, and predictive model generation (Fig. 1). insignificant variations, when various segmented volumes were For an approach to be readily transferrable to the clinic, it must be quantified (Fig. 2F). robust; our analytical pipeline demonstrated high accuracy, pre- Although in animal models, when GSEA of whole genome cision, and reliability across patients, institutions, and different expression profiles of ex vivo flow-sorted cells from the control þ þ þ vendors of MRI scanners with different magnetic field strengths (APC ve) and POSTN-knockdown (APC veGFP ve) mouse (Supplementary Table S1A and S1B). Briefly, we obtained the tumors was performed, hallmarks such as (P: outline and corresponding volume of the three GBM phenotypes 0.002; FDR Q-value: 0.001) and epithelial mesenchymal transi- (edema/invasion, enhancing active tumor and necrosis) by semi- tion (P < 0.001; FDR Q-value: 0.041), etc. were enriched in control automated consensus segmentation; consensus segmentation (i.e., high POSTN tumors; Fig. 3B). Furthermore, as suggested by produces stable results compared with single-user segmentation GSEA and confirmed by orthotopic xenograft tumor immuno- (Fig. 1A; ref. 24). The original MR images and corresponding fluorescence, the control mice from both GSC11 and 126 cell segmented masks underwent image preprocessing for extraction lines, with high POSTN expression, displayed significantly higher of stable and comparable quantitative image features (Supple- levels of Ki67 and CD31þve endothelial cells (P: <0.001) in mentary Figs. S4–S6). The inherent variability of MR signal contrast to tumors from the knockdown orthotopic mouse model intensities makes direct analysis of images obtained from different (low POSTN; Fig. 3C and D; Supplementary Fig. S7). Similar to scanners/vendors difficult; thus, MR radiomic studies to date have patients with GBM grouped according to POSTN expression mainly focused on data from single scanner and identical proto- levels, both GSC11 and 126 derived control and POSTN-knock- cols (37, 38) and only a small number of studies have utilized data down tumors developed qualitatively similar growth and phe- from multiple MR scanners/protocols (39, 40). Thus, we used a notype volumes and quantitative analyses revealed no significant brain-tissue focused normalization consisting of two steps (i) difference in volumes of contrast enhancement or peritumoral skull stripping (25) and Nyul normalization (brain-specific nor- edema/tumor invasion between the two groups of mice (Fig. 3E malization; ref. 27), followed by a patient-specific contralateral and F). hemisphere MRI normalization that allows for the extraction of stable quantitative radiomic features (Fig. 1B). Brain tissue images Radiomics predict genomic heterogeneity in GSC-derived were first obtained by excluding extra-cerebral tissues such as orthotopic tumors and GBM and is conserved across patient skull, eyeballs, fat, and skin through skull stripping, and further and mouse xenograft tumors used for normalization. Skull stripping is an important step in We then sought to determine whether radiomics could correctly studies relying on intensity levels; however, to our knowledge, it identify patients with differential expression of POSTN and the has not been used along with Nyul normalization before. The altered POSTN expression levels between control and knockdown effectiveness of Nyul normalization compared with other image tumors across the two transplanted GSC lines. Our analyses of normalization techniques has been established (28). In our study, radiomic features displayed similarities between high-POSTN we demonstrate its applicability in GBM. The combination of GBM in mice and in humans, specifically, as seen in 64 gray-level skull stripping, brain-tissue focused normalization and patient- and heterogeneity histograms (Fig. 4A). We then tested the radio- specific normalization are unique to this study and provided mic feature predictive performance by LOOCV to distinguish

www.aacrjournals.org Clin Cancer Res; 24(24) December 15, 2018 6293

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 27, 2018; DOI: 10.1158/1078-0432.CCR-17-3420

Zinn et al.

Figure 2. Qualitative and quantitative MRI parameters are unable to distinguish genomic and histologic heterogeneity in patients with GBM. Association of POSTN with distinct genomic and histologic heterogeneity in GBM. A, POSTN expression is significantly higher in mesenchymal subgroup of GBMs in comparison to proneural subgroup. Expression levels of POSTN in mesenchymal (n ¼ 124) and proneural (n ¼ 113) subgroup is shown as box plot (P value <0.0001). B, Gene set enrichment analysis shows significant enrichment of key hallmark signatures in patients with high POSTN expression. Table shows top 10 enriched gene set in patients with high POSTN expression when compared with patients with low POSTN expression. Their respective rank, net enrichment score (NES), P values, and FDR q values are shown. C, POSTN expression is higher in tumors with endothelial proliferation versus the tumors with undetectable endothelial proliferation. D, Expression levels of POSTN in tumors with endothelial proliferation (n ¼ 232) and with undetectable endothelial proliferation (n ¼ 152) is shown as box plot. , P-value: 0.0027. Qualitative and quantitative MRI parameters are unremarkable in patient with high or low POSTN expression. E, Tumors from POSTN High and POSTN Low appear qualitatively (left) and quantitatively (right) indistinguishable on representative MRI scans. F, The box plot shows quantification and comparison of volumes of necrosis, contrast enhancement, and edema (T2) in POSTN High and POSTN Low groups of patients. P values between groups were not significant.

tumors with high POSTN (control) and low POSTN (knockdown) 73.91%, specificity 78.26%; P: 0.00026; Fig. 4C) in a nonover- expression levels in two GSC lines (Fig. 4B; AUC 92.26%, sensi- lapping validation set were highly statistically significant. As a tivity 92.86%, specificity 91.67%; P: 2.68E05). Our results measure of reliability across institutions, we observed a consistent showed that unlike conventional qualitative and quantitative decrease in ICC value for these 48 features (ranging from 7% to volumetric assessments, radiomic analyses (17 radiomic features) 104%) after normalization. This decrease was significant as the correctly distinguished tumors with differential RNAi-mediated non-normalized features failed to predict POSTN levels in the POSTN status lines. Furthermore, we used radiomic analysis for patient cohort (data not shown). Furthermore, analysis with further characterization of the mouse and human tumors. radiomic features (48 radiomic features) selected for their ability Towards this, to determine the power of radiomic features to to predict POSTN levels in mouse orthotopic tumors showed predict targeted gene expression levels in tumors across species significant LOOCV of POSTN levels in the patient cohort (AUC (mouse-to-patient), we first generated and validated a radiomic 93.36%, sensitivity 82.61%, specificity 95.74%; P-value: feature-based model that successfully identified tumors based on 2.021E15; Fig. 4D; Supplementary Table S2). Finally, radio- POSTN levels in four mice cohorts across two GSC lines. As shown genomic POSTN prediction was performed in a prospective in Fig. 4B, specific radiomic features in this model significantly manner in individual patients; these radiogenomic maps showed distinguish control and knockdown xenograft tumors (AUC gene expression status (Fig. 4E). This is summarized for the patient 92.26%, sensitivity 92.86%, specificity 91.67%; P: 2.68E05). in a radiogenomic clinical report card. Accordingly, radiomic features obtained using GBM patient cohort reliably predicted POSTN status in this cohort (Supple- mentary Table S3; 48 radiomic features). Both cross-validation Discussion (AUC 99.17%, sensitivity 97.83%, specificity 97.87%; P < 2e16; In this study, for the first time to our knowledge, we demon- Supplementary Fig. S8) and prediction (AUC 76.56%, sensitivity strate a causal linkage between imaging radiomics and genomics.

6294 Clin Cancer Res; 24(24) December 15, 2018 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 27, 2018; DOI: 10.1158/1078-0432.CCR-17-3420

Validation of Radiomics and Radiogenomics

Figure 3. Qualitative and quantitative MRI parameters are unable to distinguish genomic and histologic heterogeneity in orthotopic GBM model system. A, Ex vivo validation of knockdown of POSTN in GSC11 and GSC126. Quantitative real-time PCR analysis for determination of expression levels of POSTN gene in knockdown (KD) and control tumor cells isolated from mouse brain ex vivo. Relative expression levels of POSTN after normalization to that of GAPDH in respective samples is presented (N ¼ 3). B, Gene set enrichment analysis of whole genome expression profiles of tumor cells (ex vivo) from control and KD mice. Top 10 gene set enriched in control tumor cells are shown in the table. Their respective rank, net enrichment score (NES), P values, and FDR q values are shown. C, IIHC staining of xenograft tumor sections from control and KD mice using anti-CD31 antibody. Nuclei are counterstained with hematoxylin. Respective cell lines giving rise to orthotopic tumors are labeled on the side of micrographs. Scale bar ¼ 100 mm. D, Quantification of CD31 positive cells in control and KD tumors are shown in the graph. , P-value < 0.001. E, Representative MRI scans and segmentations of GSC11 and GSC126 xenograft tumors from control (n ¼ 5, and 7, respectively) and POSTN-knockdown (KD; n ¼ 5 and 9, respectively) animals. Tumors from POSTN-wild-type (control) and POSTN-KD GSC11 and GSC126 mice appear qualitatively (top) and quantitatively (bottom) indistinguishable on T1WI and T2WI with a 7T MRI system. F, The box plot shows quantification of volumes of contrast enhancement and edema (T2) in control and POSTN-KD groups. P values between groups were not significant.

We tested our hypothesis using targeted genetic interventions. sible for maintenance of a mesenchymal program and angiogen- Herein, we demonstrate that gene expression, specifically POSTN esis in GBM (a link recently illustrated in ref. 20). These findings expression, drives a specific imaging feature phenotype. In addi- support the hypothesis that the radiogenomic functional relation- tion, we demonstrate that tumors with specific genomic expres- ships can be quite robust and may contain significant causal sion profiles harbor distinct radiomic profiles. Finally, we show linkages between MRI features and genetically driven tumor that patients harboring tumors with a distinct gene expression phenotypes. profile share significantly concordant radiomic features with their Technically underpinning this, we developed and evaluated a matching preclinical murine xenograft counterparts. novel radiomics pipeline for analysis of standard-of-care clinical Fundamental to radiogenomic analysis is the presumption that MR images, which is scalable and provides widespread applica- expression of specific sets of genes or driver mutations impact bility for high-throughput cross-vendor and software platform extractable imaging features, but this presumption has not been image processing and analysis of medical imaging applied to directly demonstrated in a highly robust model. Our results human solid tumors and preclinical tumor models. Multiple suggest that imaging features, specifically radiomic features, are innovative steps were required for this to occur and developed at least in part genetically-driven, and subtle genomic changes in throughout this process. We established and applied two unique tumor tissue are captured by radiomic changes on imaging. In steps of image normalization: first, brain-specific normalization; addition, our results show that POSTN may potentially be respon- and second, patient-specific contralateral hemisphere MRI

www.aacrjournals.org Clin Cancer Res; 24(24) December 15, 2018 6295

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 27, 2018; DOI: 10.1158/1078-0432.CCR-17-3420

Zinn et al.

Figure 4. Radiomics-based features are conserved and are predictive of POSTN levels across xenograft and human tumors. A, High POSTN tumors from patients and orthotopic xenograft mice (Control; left) show greater similarity of textural heterogeneity at 8 Gray and 64 Gray levels and are distinct from that of low POSTN tumors from patients and orthotopic xenograft mice (KD; right). B, LOOCV ROC curve depict the performance of the GSC11 and GSC126 mouse-based predictive model using 17 radiomic features to predict control and KD mice across cell lines with 92.26% accuracy and P value 2.68E05. 95% CI, sensitivity, specificity, PPV, NPV of models are shown within each plot. C, ROC curve depicts the performance of TCGA training set (n ¼ 46) based predictive model using 48 radiomic features to predict POSTN expression levels in validation set (n ¼ 47) with 76.65% accuracy. D, The LOOCV ROC curve shows the association of 17 radiomic features derived from GSC11 and GSC126 mice from panel (B) with 93 patients with endogenous high and low POSTN expression levels. E, Radiome sequencing in clinics. Radiogenomic probability maps and radiome sequencing clinical report cards for two representative patients. Summary of the radiomics-based prediction of the genomic events/status of GBM for two representative patients. The left panels show the segmented patient brain MRIs delineating the tumor. The right panels show the radiome sequencing clinical report cards summarizing the status of POSTN expression. A check mark indicates that the prediction was correct.

normalization; these allowed for robust cross-platform (ven- radiomics/tumor assessment field, although the effectiveness of dor), cross-institution patient cohort analysis, and comparison. Nyul normalization has been shown in image analysis of Genomic predictions were significant in the TCGA dataset, normal human MRIs and multiple sclerosis (28). The integra- despite these data being collated from six different medical tion of a patient-specific normalization process is new to the centers across the United States and patients having undergone radiomics field and its application in conventional imaging at imaging with MRI scanners from different vendors using dif- largehasnotbeenusedinthis setting. Thus, herein, we ferent image acquisition protocols. This unique combination of developed a novel radiomic pipeline that is agnostic to MRI normalization steps have not been previously used within the scanner vendor, institution, and protocol.

6296 Clin Cancer Res; 24(24) December 15, 2018 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 27, 2018; DOI: 10.1158/1078-0432.CCR-17-3420

Validation of Radiomics and Radiogenomics

Furthermore, we developed a pipeline that can robustly extract testing, such that noninvasive genomic profiling may enhance microscale information beyond the resolution of the naked diagnosis, help stratify patients and determine early response to human eye. As our data demonstrate, qualitative and quantitative targeted therapy; imaging radiomics also assess the entire three- (volumetric) MRI assessments did not detect any differences dimensional tumor structure and may enable monitoring of between POSTN knockdown and wild-type xenograft tumors genomic traits at a specific time for patients in whom biopsy or (Fig. 3E and F); however, radiomic analysis showed distinct surgery cannot be performed or when cost of care limits repeated radiophenotypic differences between these two groups. (See pre- genomic profiling. Hence, comprehensive radiomics can comple- and post-patient–specific normalization plots; Supplementary ment genomic information obtained via biopsy or tumor resec- Figs. S4–S6). This enhanced radiomics analysis method may tion in cancer care. It can also be anticipated that longitudinal unlock previously inaccessible data for clinicians and investiga- alterations in radiomic features can be used in combination with tors independent of where, when and how images were acquired. current response assessment criteria such as RANO, RECIST1.1, The radiomic software developed herein is scalable, potentially irRC, etc., for a more accurate and early assessment of treatment enabling multi-institutional collaborations, big data collation response. Radiomics provides significantly increased predictive and integration into clinical care, clinical trials, and research power and higher biomarker accuracy based on inherent tumor across various arenas. imaging features and pixel-based heterogeneity. POSTN, expressed in glioma cells including GSCs and secreted Despite this study being the pilot in establishing radiogenomic to in tumor microenvironment, is involved in causality within the radiogenomic literature, prospective valida- invasion and perivascular niche formation, , and mac- tion using spatially matched image-guided brain tumor biopsies rophage recruitment (19, 42). More recent studies demonstrate in patients and animal models are needed to further validate the POSTN's role in promoting glioma invasion, tumor recurrence, spatial distribution of the molecular underpinnings of imaging in and resistance to anti-angiogenic therapy (20, 43). The peritu- brain cancer. Currently, topographical spatial encoding of the moral region in gliomas harbors a mixture of edema and invasive, genomic landscape using image-guided biopsies is underway at aggressive tumor cell, a key determinant of tumor recurrence, and our institution. Another limitation of the study is that the spatial poor patient survival (44). Given POSTN's role in glioma invasion resolution (voxel size) of the human MRI is larger compared with as detailed above coupled with our previous imaging genomic the xenograft MRI; these differences could potentially affect the findings, we utilized POSTN as our gene prototype. In our study, extracted radiomic features because the distance of 1-voxel used tumors with distinct POSTN expression levels harbored distinct for the calculation of the GLCM matrix varies in size. Preproces- imaging features that were not only associated with but also sing steps that account for varying resolutions, such as upsam- predictive of expression levels in both patients and murine pling or downsampling the volume to one resolution could be models. Likewise, the preclinical model-derived radiomic features utilized; however, the goal of our study was to use the images with significantly predicted concordant gene expression in our patient the resolution dictated by their respective acquisition protocols, GBM cohort (n ¼ 93). This is a uniquely novel finding and thus no preprocessing steps were performed on xenograft models. demonstrates that detection of selected genomic traits is con- Furthermore, the results of our analyses (high accuracy, sensitiv- served in appearance on imaging across species and can be ity, and specificity) could indicate that additional preprocessing expected to further facilitate integration of radiomics and radio- steps to account for resolution differences may not be necessary genomics into clinical and co-clinical trials. For a method to be when evaluating the whole tumor compared with voxel-based translated into clinical practice robustness, generalizability and radiomic analysis. scalability of the technique are required (45). Using well-known In summary, we present a clinically applicable and meaningful robust statistical methods, we used nonoverlapping discovery and analytic imaging method for performing high-throughput image validation sets to meet and achieve high protocol standards (46). analysis that can capture key genomic events in patients with brain Giving further credence to the generalizability and scalability of cancer. Our radiomic analysis pipeline is a biologically validated our radiomic model is the fact that the pipeline is applicable test method. Furthermore, the generalizability and scalability to beyond solid tumors and to other types of imaging sequences most types of medical imaging and solid tumors can be antici- such as functional MRI (47); in a previously published study by pated and preliminary results underway at our institution using our group, we found that radiomics can detect and discriminate our pipeline are promising. It also has use beyond clinical diag- between areas of true activation in regions of eloquent cortex nosis and prognosis, as it allows for human-mouse matched versus artifact (47). coclinical trials, in-depth endpoint analysis, and upfront, nonin- The need to establish robust imaging-genomic correlations and vasive, high-resolution radiomics-based diagnostic, prognostic, the importance of radiomic tools to interrogate tissue is supported and predictive biomarker development. by multiple studies including the study by Kickingereder and colleagues who concluded that tumor volumes, perfusion, diffu- Disclosure of Potential Conflicts of Interest sion, and other MRI parameters are not sufficient for reliable and No potential conflicts of interest were disclosed. clinically meaningful predictions of molecular characteristics in patients (48). A lack of established causality as to the molecular Authors' Contributions underpinnings of imaging has been a major impediment towards Conception and design: P.O. Zinn, S.K. Singh, M.M. Luedi, A. Elakkad, general clinical acceptance of radiogenomics. However, this study G.N. Fuller, J.F. de Groot, R.R. Colen demonstrates that radiomic assessment can predict genomically Development of methodology: P.O. Zinn, S.K. Singh, A. Kotrotsou, distinct tumors with high sensitivity, specificity, accuracy, and M.M. Luedi, A. Elakkad, N. Elshafeey, J. Gumin, R.R. Colen Acquisition of data (provided animals, acquired and managed patients, evaluate pertinent genomic alterations towards providing provided facilities, etc.): P.O. Zinn, S.K. Singh, A. Kotrotsou, I. Hassan, detailed prediction maps of the molecular GBM landscape. This G. Thomas, M.M. Luedi, A. Elakkad, N. Elshafeey, T. Idris, J.F. de Groot, method will not replace but complement conventional genomic E.P. Sulman, F.F. Lang, R.R. Colen

www.aacrjournals.org Clin Cancer Res; 24(24) December 15, 2018 6297

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 27, 2018; DOI: 10.1158/1078-0432.CCR-17-3420

Zinn et al.

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, funding. Radiological Society of North America scholar grant (RSCH11506). computational analysis): P.O. Zinn, S.K. Singh, A. Kotrotsou, I. Hassan, Cancer Prevention and Research Institute of Texas (CPRIT) grant (RP160150; to M.M. Luedi, N. Elshafeey, T. Idris, J.F. de Groot, V. Baladandayuthapani, R.R. Colen). This work was also supported by the NIH R25 Baylor College of A.J. Kumar, R. Sawaya, D. Piwnica-Worms, R.R. Colen Medicine, Department of Neurosurgery Research Grant and Neurosurgery Writing, review, and/or revision of the manuscript: P.O. Zinn, S.K. Singh, Research And Education Foundation (NREF; 338703; to P.O. Zinn). Further A. Kotrotsou, I. Hassan, M.M. Luedi, J. Mosley, G.N. Fuller, J.F. de Groot, support includes MD Anderson's Cancer Center support grant (CA016672); A.J. Kumar, R. Sawaya, D. Piwnica-Worms, R.R. Colen NCI (P50 CA127001; animal core and biospecimen core), the Elias Family Administrative, technical, or material support (i.e., reporting or organizing Fund, and the Broach Foundation (to F.F. Lang). data, constructing databases): P.O. Zinn, I. Hassan, M.M. Luedi, J. Mosley, R.R. Colen Study supervision: R.R. Colen The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement Acknowledgments in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. We thank Joseph Munch for editorial help and V.L. Brandt for shepherding the manuscript through multiple drafts. We also thank the NCI and the TCGA/ TCIA Glioma Phenotype Research Group. This work was supported by John S. Received November 23, 2017; revised March 31, 2018; accepted July 24, 2018; Dunn Sr. Distinguished Chair in Diagnostic Imaging fund, MDACC startup published first July 27, 2018.

References 1. Eisenhauer E, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. 16. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al. New response evaluation criteria in solid tumours: revised RECIST MGMT Gene silencing and benefit from temozolomide in glioblastoma. guideline (version 1.1). Eur J Cancer 2009;45:228–47. N Engl J Med 2005;352:997–1003. 2. Nishino M, Giobbie-Hurder A, Gargano M, Suda M, Ramaiya NH, Hodi FS. 17. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Developing a common language for tumor response to immunotherapy: et al. Radiotherapy plus concomitant and adjuvant temozolomide for immune-related response criteria using unidimensional measurements. glioblastoma. N Engl J Med 2005;352:987–96. Clin Cancer Res 2013;19:3936–43. 18. Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, et al. 3. Wen PY, Macdonald DR, Reardon DA, Cloughesy TF, Sorensen AG, Galanis Integrated genomic analysis identifies clinically relevant subtypes of glio- E, et al. Updated response assessment criteria for high-grade gliomas: blastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and response assessment in neuro-oncology working group. J Clin Oncol NF1. Cancer Cell 2010;17:98–110. 2010;28:1963–72. 19. Zhou W, Ke SQ, Huang Z, Flavahan W, Fang X, Paul J, et al. Periostin 4. Wolchok JD, Hoos A, O'Day S, Weber JS, Hamid O, Lebbe C, et al. secreted by glioblastoma stem cells recruits M2 tumour-associated macro- Guidelines for the evaluation of immune therapy activity in solid tumors: phages and promotes malignant growth. Nat Cell Biol 2015;17:170–82. immune-related response criteria. Clin Cancer Res 2009;15:7412–20. 20. Park SY, Piao Y, Jeong KJ, Dong J, de Groot JF. Periostin (POSTN) regulates 5. Aerts HJ, Velazquez ER, Leijenaar RT, Parmar C, Grossmann P, Carvalho S, tumor resistance to antiangiogenic therapy in glioma models. Mol Cancer et al. Decoding tumour phenotype by noninvasive imaging using a quan- Ther 2016;15:2187–97. titative radiomics approach. Nat Commun 2014;5:4006. 21. Archip N, Jolesz FA, Warfield SK. A validation framework for brain tumor 6. Lambin P, Rios-Velazquez E, Leijenaar R, Carvalho S, van Stiphout RG, segmentation. Acad Radiol 2007;14:1242–51. Granton P, et al. Radiomics: extracting more information from medical 22. Gering DT, Nabavi A, Kikinis R, Hata N, O'Donnell LJ, Grimson WE, images using advanced feature analysis. Eur J Cancer 2012;48:441–6. et al. An integrated visualization system for surgical planning and 7. Li H, Zhu Y, Burnside ES, Drukker K, Hoadley KA, Fan C, et al. MR imaging guidance using image fusion and an open MR. J Magn Reson Imaging radiomics signatures for predicting the risk of breast cancer recurrence as 2001;13:967–75. given by research versions of MammaPrint, Oncotype DX, and PAM50 gene 23. Pichon E, Tannenbaum A, Kikinis R. A statistically based flow for image assays. Radiology 2016;281:382–91. segmentation. Med Image Anal 2004;8:267–74. 8. Castellano G, Bonilha L, Li LM, Cendes F. Texture analysis of medical 24. Zinn PO, Singh SK, Kotrotsou A, Abrol S, Thomas G, Mosley J, et al. Distinct images. Clin Radiol 2004;59:1061–9. radiomic phenotypes define glioblastoma TP53-PTEN-EGFR mutational 9. Davnall F, Yip CS, Ljungqvist G, Selmi M, Ng F, Sanghera B, et al. landscape. Neurosurgery 2017;64(CN_suppl_1):203–10. Assessment of tumor heterogeneity: an emerging imaging tool for clinical 25. Jenkinson M, Pechaud M, Smith S. BET2: MR-based estimation of brain, practice? Insights Imaging 2012;3:573–89. skull and scalp surfaces. In: Proceedings of the Eleventh Annual Meeting of 10. Diehn M, Nardini C, Wang DS, McGovern S, Jayaraman M, Liang Y, et al. the Organisation for Human Brain Mapping; 2005; Toronto, Canada. Identification of noninvasive imaging surrogates for brain tumor gene- Minneapolis (MN): OHBM; 2005. expression modules. Proc Natl Acad Sci U S A 2008;105:5213–8. 26. Smith SM. Fast robust automated brain extraction. Hum Brain Mapp 11. Preusser M, de Ribaupierre S, Wohrer€ A, Erridge SC, Hegi M, Weller M, et al. 2002;17:143–55. Current concepts and management of glioblastoma. Ann Neurol 2011; 27. Nyul LG, Udupa JK, Zhang X. New variants of a method of MRI scale 70:9–21. standardization. IEEE Trans Med Imaging 2000;19:143–50. 12. Wu W, Parmar C, Grossmann P, Quackenbush J, Lambin P, Bussink J, et al. 28. Shah M, Xiao Y, Subbanna N, Francis S, Arnold DL, Collins DL, et al. Exploratory study to identify radiomics classifiers for lung cancer histology. Evaluating intensity normalization on MRIs of human brain with multiple Front Oncol 2016;6:71. sclerosis. Med Image Anal 2011;15:267–82. 13. Zinn PO, Mahajan B, Sathyan P, Singh SK, Majumder S, Jolesz FA, et al. 29. Shinohara RT, Sweeney EM, Goldsmith J, Shiee N, Mateen FJ, Calabresi PA, Radiogenomic mapping of edema/cellular invasion MRI-phenotypes in et al. Statistical normalization techniques for magnetic resonance imaging. glioblastoma multiforme. PLoS One 2011;6:e25451. Neuroimage Clin 2014;6:9–19. 14. Joo KM, Kim J, Jin J, Kim M, Seol HJ, Muradov J, et al. Patient-specific 30. Haralick RM, Shanmugam K, Dinstein IH. Textural features for image orthotopic glioblastoma xenograft models recapitulate the histopa- classification. IEEE Tran Syst Man Cybern 1973;3:610–21. thology and biology of human glioblastomasinsitu.CellRep2013; 31. Papoulis A. Probability, random variables, and stochastic processes. 3rd ed. 3:260–73. McGraw-Hill series in electrical engineering Communications and signal 15. Panth KM, Leijenaar RT, Carvalho S, Lieuwes NG, Yaromina A, Dubois processing. New York, NY: McGraw-Hill; 1991. p. 666. L, et al. Is there a causal relationship between genetic changes and 32. Pratt WK. Digital image processing. 2nd ed. New York, NY: Wiley; 1991. radiomics-based image features? An in vivo preclinical experiment with p. 698. doxycycline inducible GADD34 tumor cells. Radiother Oncol 2015; 33. Clausi DA. An analysis of co-occurrence texture statistics as a function of 116:462–6. grey level quantization. Can J Remote Sens 2014;28:45–62.

6298 Clin Cancer Res; 24(24) December 15, 2018 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 27, 2018; DOI: 10.1158/1078-0432.CCR-17-3420

Validation of Radiomics and Radiogenomics

34. Soh LK, Tsatsoulis C. Texture analysis of SAR sea ice imagery using gray 41. Gutman DA, Cobb J, Somanna D, Park Y, Wang F, Kurc T, et al. Cancer level co-occurrence matrices. IEEE Trans Geosci Remote Sens 1999; Digital Slide Archive: an informatics resource to support integrated in silico 37:780–95. analysis of TCGA pathology data. J Am Med Inform Assoc 2013;20:1091–8. 35. Tibshirani R. Regression shrinkage and selection via the lasso. J R Stat Soc 42. Malanchi I, Santamaria-Martínez A, Susanto E, Peng H, Lehr HA, Delaloye Series B Methodol 1996;58:267–88. JF, et al. Interactions between cancer stem cells and their niche govern 36. Chen T, Guestrin C. Xgboost: A scalable tree boosting system. In: Proceed- metastatic colonization. Nature 2012;481:85–9. ings of the 22nd ACM SIGKDD International Conference on Knowledge 43. Mikheev AM, Mikheeva SA, Trister AD, Tokita MJ, Emerson SN, Parada CA, Discovery and Data Mining; 2016 Aug 13–17; San Francisco, CA. New et al. Periostin is a novel therapeutic target that predicts and regulates York, NY: ACM; 2016. p. 785–94. glioma malignancy. Neuro Oncol 2015;17:372–82. 37. HuLS,NingS,EschbacherJM,GawN,DueckAC,SmithKA,etal.Multi- 44. Wangaryattawanich P, Hatami M, Wang J, Thomas G, Flanders A, Kirby J, parametric MRI and texture analysis to visualize spatial histologic et al. Multicenter imaging outcomes study of The Cancer Genome Atlas heterogeneity and tumor extent in glioblastoma. PLoS One 2015;10: glioblastoma patient cohort: imaging predictors of overall and progres- e0141506. sion-free survival. Neuro Oncol 2015;17:1525–37. 38. Wiestler B, Kluge A, Lukas M, Gempt J, Ringel F, Schlegel J, et al. Multi- 45. Kumar V, Gu Y, Basu S, Berglund A, Eschrich SA, Schabath MB, et al. parametric MRI-based differentiation of WHO grade II/III glioma and Radiomics: the process and the challenges. Magn Reson Imaging 2012; WHO grade IV glioblastoma. Sci Rep 2016;6:35142. 30:1234–48. 39. Prasanna P, Patel J, Partovi S, Madabhushi A, Tiwari P. Radiomic 46. Bleeker SE, Moll HA, Steyerberg EW, Donders AR, Derksen-Lubsen G, features from the peritumoral brain parenchyma on treatment-naive Grobbee DE, et al. External validation is necessary in prediction research: a multi-parametric MR imaging predict long versus short-term survival in clinical example. J Clin Epidemiol 2003;56:826–32. glioblastoma multiforme: preliminary findings. Eur Radiol 2017;27: 47. Hassan I, Kotrotsou A, Bakhtiari AS, Thomas GA, Weinberg JS, Kumar AJ, 4188–97. et al. Radiomic texture analysis mapping predicts areas of true functional 40. Upadhaya T, Morvan Y, Stindel E, Le Reste P-J, Hatt M, et al. Prognosis MRI activity. Sci Rep 2016;6:25295. classification in glioblastoma multiforme using multimodal MRI derived 48. Kickingereder P, Burth S, Wick A, Gotz€ M, Eidel O, Schlemmer HP, et al. heterogeneity textural features: impact of pre-processing choices. In: Pro- Radiomic profiling of glioblastoma: identifying an imaging predictor of ceedings of Medical Imaging 2016: Computer-Aided Diagnosis; 2016 Mar patient survival with improved performance over established clinical and 24; San Diego, CA. Bellingham, WA: SPIE; 2016 radiologic risk models. Radiology 2016;280:880–9.

www.aacrjournals.org Clin Cancer Res; 24(24) December 15, 2018 6299

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst July 27, 2018; DOI: 10.1158/1078-0432.CCR-17-3420

A Coclinical Radiogenomic Validation Study: Conserved Magnetic Resonance Radiomic Appearance of Periostin-Expressing Glioblastoma in Patients and Xenograft Models

Pascal O. Zinn, Sanjay K. Singh, Aikaterini Kotrotsou, et al.

Clin Cancer Res 2018;24:6288-6299. Published OnlineFirst July 27, 2018.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-17-3420

Supplementary Access the most recent supplemental material at: Material http://clincancerres.aacrjournals.org/content/suppl/2018/07/27/1078-0432.CCR-17-3420.DC1

Cited articles This article cites 43 articles, 5 of which you can access for free at: http://clincancerres.aacrjournals.org/content/24/24/6288.full#ref-list-1

Citing articles This article has been cited by 5 HighWire-hosted articles. Access the articles at: http://clincancerres.aacrjournals.org/content/24/24/6288.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/24/24/6288. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research.