Radiomics and Cancers

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Methods and Technologies Development".

Deadline for manuscript submissions: closed (30 September 2020) | Viewed by 88152

Special Issue Editor


E-Mail Website
Guest Editor
Department of Radiology and Nuclear Medicine, Otto-von-Guericke University Magdeburg, Leipziger Str 44, 39112 Magdeburg, Germany
Interests: radiomics; oncology; histopathology; survival
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Different radiological imaging methods are used in oncology to investigate different regions of the body for detection of primary tumors and metastatic spread, i.e., for tumor staging. Nowadays, imaging can also characterize several lesions and predict their histopathological features. Furthermore, imaging can predict tumor behavior and prognosis.

Radiomics represents a novel imaging analysis technique, which consists of extracting numerous quantitative features using automated or semi-automated software. Radiomics is based on the hypothesis that mineable data can be extracted from medical images and provide additional information on gene protein and tumor phenotype, which can then be used for patient care.

The purpose of the present Special Issue is to analyze the possible relationships between different histopathological features and radiomics parameters in several malignancies.

Dr. Alexey Surov
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • radiomics
  • imaging
  • oncology
  • histopathology
  • survival

Published Papers (24 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review, Other

14 pages, 803 KiB  
Article
Prior PSMA PET-CT Imaging and Hounsfield Unit Impact on Tumor Yield and Success of Molecular Analyses from Bone Biopsies in Metastatic Prostate Cancer
by Minke Smits, Kamer Ekici, Samhita Pamidimarri Naga, Inge M. van Oort, Michiel J. P. Sedelaar, Jack A. Schalken, James Nagarajah, Tom W. J. Scheenen, Winald R. Gerritsen, Jurgen J. Fütterer and Niven Mehra
Cancers 2020, 12(12), 3756; https://doi.org/10.3390/cancers12123756 - 14 Dec 2020
Cited by 4 | Viewed by 1754
Abstract
Developing and optimizing targeted therapies in metastatic castration-resistant prostate cancer (mCRPC) necessitates molecular characterization. Obtaining sufficient tumor material for molecular characterization has been challenging. We aimed to identify clinical and imaging variables of imaging-guided bone biopsies in metastatic prostate cancer patients that associate [...] Read more.
Developing and optimizing targeted therapies in metastatic castration-resistant prostate cancer (mCRPC) necessitates molecular characterization. Obtaining sufficient tumor material for molecular characterization has been challenging. We aimed to identify clinical and imaging variables of imaging-guided bone biopsies in metastatic prostate cancer patients that associate with tumor yield and success in obtaining molecular results, and to design a predictive model: Clinical and imaging data were collected retrospectively from patients with prostate cancer who underwent a bone biopsy for histological and molecular characterization. Clinical characteristics, imaging modalities and imaging variables, were associated with successful biopsy results. In our study, we included a total of 110 bone biopsies. Histological conformation was possible in 84 of all biopsies, of which, in 73 of the 84, successful molecular characterization was performed. Prior use of PSMA PET-CT resulted in higher success rates in histological and molecular successful biopsies compared to CT or MRI. Evaluation of spine biopsies showed more often successful results compared to other locations for both histological and molecular biopsies (p = 0.027 and p = 0.012, respectively). Low Hounsfield units (HUs) and deviation (Dev), taken at CT-guidance, were associated with histological successful biopsies (p = 0.025 and p = 0.023, respectively) and with molecular successful biopsies (p = 0.010 and p = 0.006, respectively). A prediction tool combining low HUs and low Dev resulted in significantly more successful biopsies, histological and molecular (p = 0.023 and p = 0.007, respectively). Based on these results, we concluded that site selection for metastatic tissue biopsies with prior PSMA PET-CT imaging improves the chance of a successful biopsy. Further optimization can be achieved at CT-guidance, by selection of low HU and low Dev lesions. A prediction tool is provided to increase the success rate of bone biopsies in mCRPC patients, which can easily be implemented in daily practice. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

17 pages, 6709 KiB  
Article
Identifying Cross-Scale Associations between Radiomic and Pathomic Signatures of Non-Small Cell Lung Cancer Subtypes: Preliminary Results
by Charlems Alvarez-Jimenez, Alvaro A. Sandino, Prateek Prasanna, Amit Gupta, Satish E. Viswanath and Eduardo Romero
Cancers 2020, 12(12), 3663; https://doi.org/10.3390/cancers12123663 - 07 Dec 2020
Cited by 34 | Viewed by 3655
Abstract
(1) Background: Despite the complementarity between radiology and histopathology, both from a diagnostic and a prognostic perspective, quantitative analyses of these modalities are usually performed in disconnected silos. This work presents initial results for differentiating two major non-small cell lung cancer (NSCLC) subtypes [...] Read more.
(1) Background: Despite the complementarity between radiology and histopathology, both from a diagnostic and a prognostic perspective, quantitative analyses of these modalities are usually performed in disconnected silos. This work presents initial results for differentiating two major non-small cell lung cancer (NSCLC) subtypes by exploring cross-scale associations between Computed Tomography (CT) images and corresponding digitized pathology images. (2) Methods: The analysis comprised three phases, (i) a multi-resolution cell density quantification to identify discriminant pathomic patterns for differentiating adenocarcinoma (ADC) and squamous cell carcinoma (SCC), (ii) radiomic characterization of CT images by using Haralick descriptors to quantify tumor textural heterogeneity as represented by gray-level co-occurrences to discriminate the two pathological subtypes, and (iii) quantitative correlation analysis between the multi-modal features to identify potential associations between them. This analysis was carried out using two publicly available digitized pathology databases (117 cases from TCGA and 54 cases from CPTAC) and a public radiological collection of CT images (101 cases from NSCLC-R). (3) Results: The top-ranked cell density pathomic features from the histopathology analysis were correlation, contrast, homogeneity, sum of entropy and difference of variance; which yielded a cross-validated AUC of 0.72 ± 0.02 on the training set (CPTAC) and hold-out validation AUC of 0.77 on the testing set (TCGA). Top-ranked co-occurrence radiomic features within NSCLC-R were contrast, correlation and sum of entropy which yielded a cross-validated AUC of 0.72 ± 0.01. Preliminary but significant cross-scale associations were identified between cell density statistics and CT intensity values using matched specimens available in the TCGA cohort, which were used to significantly improve the overall discriminatory performance of radiomic features in differentiating NSCLC subtypes (AUC = 0.78 ± 0.01). (4) Conclusions: Initial results suggest that cross-scale associations may exist between digital pathology and CT imaging which can be used to identify relevant radiomic and histopathology features to accurately distinguish lung adenocarcinomas from squamous cell carcinomas. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Graphical abstract

19 pages, 2374 KiB  
Article
CT-Based Radiomics Analysis to Predict Malignancy in Patients with Intraductal Papillary Mucinous Neoplasm (IPMN) of the Pancreas
by David Tobaly, Joao Santinha, Riccardo Sartoris, Marco Dioguardi Burgio, Celso Matos, Jérôme Cros, Anne Couvelard, Vinciane Rebours, Alain Sauvanet, Maxime Ronot, Nikolaos Papanikolaou and Valérie Vilgrain
Cancers 2020, 12(11), 3089; https://doi.org/10.3390/cancers12113089 - 23 Oct 2020
Cited by 33 | Viewed by 3248
Abstract
To assess the performance of CT-based radiomics analysis in differentiating benign from malignant intraductal papillary mucinous neoplasms of the pancreas (IPMN), preoperative scans of 408 resected patients with IPMN were retrospectively analyzed. IPMNs were classified as benign (low-grade dysplasia, n = 181), or [...] Read more.
To assess the performance of CT-based radiomics analysis in differentiating benign from malignant intraductal papillary mucinous neoplasms of the pancreas (IPMN), preoperative scans of 408 resected patients with IPMN were retrospectively analyzed. IPMNs were classified as benign (low-grade dysplasia, n = 181), or malignant (high grade, n = 128, and invasive, n = 99). Clinicobiological data were reported. Patients were divided into a training cohort (TC) of 296 patients and an external validation cohort (EVC) of 112 patients. After semi-automatic tumor segmentation, PyRadiomics was used to extract radiomics features. A multivariate model was developed using a logistic regression approach. In the training cohort, 85/107 radiomics features were significantly different between patients with benign and malignant IPMNs. Unsupervised clustering analysis revealed four distinct clusters of patients with similar radiomics features patterns with malignancy as the most significant association. The multivariate model differentiated benign from malignant tumors in TC with an area under the ROC curve (AUC) of 0.84, sensitivity (Se) of 0.82, specificity (Spe) of 0.74, and in EVC with an AUC of 0.71, Se of 0.69, Spe of 0.57. This large study confirms the high diagnostic performance of preoperative CT-based radiomics analysis to differentiate between benign from malignant IPMNs. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

16 pages, 2766 KiB  
Article
Radiomic Immunophenotyping of GSEA-Assessed Immunophenotypes of Glioblastoma and Its Implications for Prognosis: A Feasibility Study
by Justin Bo-Kai Hsu, Gilbert Aaron Lee, Tzu-Hao Chang, Shiu-Wen Huang, Nguyen Quoc Khanh Le, Yung-Chieh Chen, Duen-Pang Kuo, Yi-Tien Li and Cheng-Yu Chen
Cancers 2020, 12(10), 3039; https://doi.org/10.3390/cancers12103039 - 19 Oct 2020
Cited by 21 | Viewed by 2818
Abstract
Characterization of immunophenotypes in glioblastoma (GBM) is important for therapeutic stratification and helps predict treatment response and prognosis. Radiomics can be used to predict molecular subtypes and gene expression levels. However, whether radiomics aids immunophenotyping prediction is still unknown. In this study, to [...] Read more.
Characterization of immunophenotypes in glioblastoma (GBM) is important for therapeutic stratification and helps predict treatment response and prognosis. Radiomics can be used to predict molecular subtypes and gene expression levels. However, whether radiomics aids immunophenotyping prediction is still unknown. In this study, to classify immunophenotypes in patients with GBM, we developed machine learning-based magnetic resonance (MR) radiomic models to evaluate the enrichment levels of four immune subsets: Cytotoxic T lymphocytes (CTLs), activated dendritic cells, regulatory T cells (Tregs), and myeloid-derived suppressor cells (MDSCs). Independent testing data and the leave-one-out cross-validation method were used to evaluate model effectiveness and model performance, respectively. We identified five immunophenotypes (G1 to G5) based on the enrichment level for the four immune subsets. G2 had the worst prognosis and comprised highly enriched MDSCs and lowly enriched CTLs. G3 had the best prognosis and comprised lowly enriched MDSCs and Tregs and highly enriched CTLs. The average accuracy of T1-weighted contrasted MR radiomics models of the enrichment level for the four immune subsets reached 79% and predicted G2, G3, and the “immune-cold” phenotype (G1) according to our radiomics models. Our radiomic immunophenotyping models feasibly characterize the immunophenotypes of GBM and can predict patient prognosis. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

13 pages, 2103 KiB  
Article
Radiomic Features and Machine Learning for the Discrimination of Renal Tumor Histological Subtypes: A Pragmatic Study Using Clinical-Routine Computed Tomography
by Johannes Uhlig, Andreas Leha, Laura M. Delonge, Anna-Maria Haack, Brian Shuch, Hyun S. Kim, Felix Bremmer, Lutz Trojan, Joachim Lotz and Annemarie Uhlig
Cancers 2020, 12(10), 3010; https://doi.org/10.3390/cancers12103010 - 16 Oct 2020
Cited by 20 | Viewed by 2648
Abstract
This study evaluates the diagnostic performance of radiomic features and machine learning algorithms for renal tumor subtype assessment in venous computed tomography (CT) studies from clinical routine. Patients undergoing surgical resection and histopathological assessment of renal tumors at a tertiary referral center between [...] Read more.
This study evaluates the diagnostic performance of radiomic features and machine learning algorithms for renal tumor subtype assessment in venous computed tomography (CT) studies from clinical routine. Patients undergoing surgical resection and histopathological assessment of renal tumors at a tertiary referral center between 2012 and 2019 were included. Preoperative venous-phase CTs from multiple referring imaging centers were segmented, and standardized radiomic features extracted. After preprocessing, class imbalance handling, and feature selection, machine learning algorithms were used to predict renal tumor subtypes using 10-fold cross validation, assessed as multiclass area under the curve (AUC). In total, n = 201 patients were included (73.7% male; mean age 66 ± 11 years), with n = 131 clear cell renal cell carcinomas (ccRCC), n = 29 papillary RCC, n = 11 chromophobe RCC, n = 16 oncocytomas, and n = 14 angiomyolipomas (AML). An extreme gradient boosting algorithm demonstrated the highest accuracy (multiclass area under the curve (AUC) = 0.72). The worst discrimination was evident for oncocytomas vs. AML and oncocytomas vs. chromophobe RCC (AUC = 0.55 and AUC = 0.45, respectively). In sensitivity analyses excluding oncocytomas, a random forest algorithm showed the highest accuracy, with multiclass AUC = 0.78. Radiomic feature analyses from venous-phase CT acquired in clinical practice with subsequent machine learning can discriminate renal tumor subtypes with moderate accuracy. The classification of oncocytomas seems to be the most complex with the lowest accuracy. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

20 pages, 3321 KiB  
Article
Baseline MRI-Radiomics Can Predict Overall Survival in Non-Endemic EBV-Related Nasopharyngeal Carcinoma Patients
by Marco Bologna, Valentina Corino, Giuseppina Calareso, Chiara Tenconi, Salvatore Alfieri, Nicola Alessandro Iacovelli, Anna Cavallo, Stefano Cavalieri, Laura Locati, Paolo Bossi, Domenico Attilio Romanello, Rossana Ingargiola, Tiziana Rancati, Emanuele Pignoli, Silvana Sdao, Mattia Pecorilla, Nadia Facchinetti, Annalisa Trama, Lisa Licitra, Luca Mainardi and Ester Orlandiadd Show full author list remove Hide full author list
Cancers 2020, 12(10), 2958; https://doi.org/10.3390/cancers12102958 - 13 Oct 2020
Cited by 33 | Viewed by 2381
Abstract
Advanced stage nasopharyngeal cancer (NPC) shows highly variable treatment outcomes, suggesting the need for independent prognostic factors. This study aims at developing a magnetic resonance imaging (MRI)-based radiomic signature as a prognostic marker for different clinical endpoints in NPC patients from non-endemic areas. [...] Read more.
Advanced stage nasopharyngeal cancer (NPC) shows highly variable treatment outcomes, suggesting the need for independent prognostic factors. This study aims at developing a magnetic resonance imaging (MRI)-based radiomic signature as a prognostic marker for different clinical endpoints in NPC patients from non-endemic areas. A total 136 patients with advanced NPC and available MRI imaging (T1-weighted and T2-weighted) were selected. For each patient, 2144 radiomic features were extracted from the main tumor and largest lymph node. A multivariate Cox regression model was trained on a subset of features to obtain a radiomic signature for overall survival (OS), which was also applied for the prognosis of other clinical endpoints. Validation was performed using 10-fold cross-validation. The added prognostic value of the radiomic features to clinical features and volume was also evaluated. The radiomics-based signature had good prognostic power for OS and loco-regional recurrence-free survival (LRFS), with C-index of 0.68 and 0.72, respectively. In all the cases, the addition of radiomics to clinical features improved the prognostic performance. Radiomic features can provide independent prognostic information in NPC patients from non-endemic areas. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

23 pages, 4823 KiB  
Article
Integrated Multiparametric Radiomics and Informatics System for Characterizing Breast Tumor Characteristics with the OncotypeDX Gene Assay
by Michael A. Jacobs, Christopher B. Umbricht, Vishwa S. Parekh, Riham H. El Khouli, Leslie Cope, Katarzyna J. Macura, Susan Harvey and Antonio C. Wolff
Cancers 2020, 12(10), 2772; https://doi.org/10.3390/cancers12102772 - 27 Sep 2020
Cited by 15 | Viewed by 3480
Abstract
Optimal use of multiparametric magnetic resonance imaging (mpMRI) can identify key MRI parameters and provide unique tissue signatures defining phenotypes of breast cancer. We have developed and implemented a new machine-learning informatic system, termed Informatics Radiomics Integration System (IRIS) that integrates clinical variables, [...] Read more.
Optimal use of multiparametric magnetic resonance imaging (mpMRI) can identify key MRI parameters and provide unique tissue signatures defining phenotypes of breast cancer. We have developed and implemented a new machine-learning informatic system, termed Informatics Radiomics Integration System (IRIS) that integrates clinical variables, derived from imaging and electronic medical health records (EHR) with multiparametric radiomics (mpRad) for identifying potential risk of local or systemic recurrence in breast cancer patients. We tested the model in patients (n = 80) who had Estrogen Receptor positive disease and underwent OncotypeDX gene testing, radiomic analysis, and breast mpMRI. The IRIS method was trained using the mpMRI, clinical, pathologic, and radiomic descriptors for prediction of the OncotypeDX risk score. The trained mpRad IRIS model had a 95% and specificity was 83% with an Area Under the Curve (AUC) of 0.89 for classifying low risk patients from the intermediate and high-risk groups. The lesion size was larger for the high-risk group (2.9 ± 1.7 mm) and lower for both low risk (1.9 ± 1.3 mm) and intermediate risk (1.7 ± 1.4 mm) groups. The lesion apparent diffusion coefficient (ADC) map values for high- and intermediate-risk groups were significantly (p < 0.05) lower than the low-risk group (1.14 vs. 1.49 × 10−3 mm2/s). These initial studies provide deeper insight into the clinical, pathological, quantitative imaging, and radiomic features, and provide the foundation to relate these features to the assessment of treatment response for improved personalized medicine. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

11 pages, 1231 KiB  
Article
Computer Extracted Features from Initial H&E Tissue Biopsies Predict Disease Progression for Prostate Cancer Patients on Active Surveillance
by Sacheth Chandramouli, Patrick Leo, George Lee, Robin Elliott, Christine Davis, Guangjing Zhu, Pingfu Fu, Jonathan I. Epstein, Robert Veltri and Anant Madabhushi
Cancers 2020, 12(9), 2708; https://doi.org/10.3390/cancers12092708 - 21 Sep 2020
Cited by 15 | Viewed by 3321
Abstract
In this work, we assessed the ability of computerized features of nuclear morphology from diagnostic biopsy images to predict prostate cancer (CaP) progression in active surveillance (AS) patients. Improved risk characterization of AS patients could reduce over-testing of low-risk patients while directing high-risk [...] Read more.
In this work, we assessed the ability of computerized features of nuclear morphology from diagnostic biopsy images to predict prostate cancer (CaP) progression in active surveillance (AS) patients. Improved risk characterization of AS patients could reduce over-testing of low-risk patients while directing high-risk patients to therapy. A total of 191 (125 progressors, 66 non-progressors) AS patients from a single site were identified using The Johns Hopkins University’s (JHU) AS-eligibility criteria. Progression was determined by pathologists at JHU. 30 progressors and 30 non-progressors were randomly selected to create the training cohort D1 (n = 60). The remaining patients comprised the validation cohort D2 (n = 131). Digitized Hematoxylin & Eosin (H&E) biopsies were annotated by a pathologist for CaP regions. Nuclei within the cancer regions were segmented using a watershed method and 216 nuclear features describing position, shape, orientation, and clustering were extracted. Six features associated with disease progression were identified using D1 and then used to train a machine learning classifier. The classifier was validated on D2. The classifier was further compared on a subset of D2 (n = 47) against pro-PSA, an isoform of prostate specific antigen (PSA) more linked with CaP, in predicting progression. Performance was evaluated with area under the curve (AUC). A combination of nuclear spatial arrangement, shape, and disorder features were associated with progression. The classifier using these features yielded an AUC of 0.75 in D2. On the 47 patient subset with pro-PSA measurements, the classifier yielded an AUC of 0.79 compared to an AUC of 0.42 for pro-PSA. Nuclear morphometric features from digitized H&E biopsies predicted progression in AS patients. This may be useful for identifying AS-eligible patients who could benefit from immediate curative therapy. However, additional multi-site validation is needed. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

13 pages, 12752 KiB  
Article
Analysis of Bone Scans in Various Tumor Entities Using a Deep-Learning-Based Artificial Neural Network Algorithm—Evaluation of Diagnostic Performance
by Jan Wuestemann, Sebastian Hupfeld, Dennis Kupitz, Philipp Genseke, Simone Schenke, Maciej Pech, Michael C. Kreissl and Oliver S. Grosser
Cancers 2020, 12(9), 2654; https://doi.org/10.3390/cancers12092654 - 17 Sep 2020
Cited by 19 | Viewed by 3979
Abstract
The bone scan index (BSI), initially introduced for metastatic prostate cancer, quantifies the osseous tumor load from planar bone scans. Following the basic idea of radiomics, this method incorporates specific deep-learning techniques (artificial neural network) in its development to provide automatic calculation, feature [...] Read more.
The bone scan index (BSI), initially introduced for metastatic prostate cancer, quantifies the osseous tumor load from planar bone scans. Following the basic idea of radiomics, this method incorporates specific deep-learning techniques (artificial neural network) in its development to provide automatic calculation, feature extraction, and diagnostic support. As its performance in tumor entities, not including prostate cancer, remains unclear, our aim was to obtain more data about this aspect. The results of BSI evaluation of bone scans from 951 consecutive patients with different tumors were retrospectively compared to clinical reports (bone metastases, yes/no). Statistical analysis included entity-specific receiver operating characteristics to determine optimized BSI cut-off values. In addition to prostate cancer (cut-off = 0.27%, sensitivity (SN) = 87%, specificity (SP) = 99%), the algorithm used provided comparable results for breast cancer (cut-off 0.18%, SN = 83%, SP = 87%) and colorectal cancer (cut-off = 0.10%, SN = 100%, SP = 90%). Worse performance was observed for lung cancer (cut-off = 0.06%, SN = 63%, SP = 70%) and renal cell carcinoma (cut-off = 0.30%, SN = 75%, SP = 84%). The algorithm did not perform satisfactorily in melanoma (SN = 60%). For most entities, a high negative predictive value (NPV ≥ 87.5%, melanoma 80%) was determined, whereas positive predictive value (PPV) was clinically not applicable. Automatically determined BSI showed good sensitivity and specificity in prostate cancer and various other entities. Particularly, the high NPV encourages applying BSI as a tool for computer-aided diagnostic in various tumor entities. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

12 pages, 822 KiB  
Article
Multi-Parametric Deep Learning Model for Prediction of Overall Survival after Postoperative Concurrent Chemoradiotherapy in Glioblastoma Patients
by Han Gyul Yoon, Wonjoong Cheon, Sang Woon Jeong, Hye Seung Kim, Kyunga Kim, Heerim Nam, Youngyih Han and Do Hoon Lim
Cancers 2020, 12(8), 2284; https://doi.org/10.3390/cancers12082284 - 14 Aug 2020
Cited by 18 | Viewed by 3105
Abstract
This study aimed to investigate the performance of a deep learning-based survival-prediction model, which predicts the overall survival (OS) time of glioblastoma patients who have received surgery followed by concurrent chemoradiotherapy (CCRT). The medical records of glioblastoma patients who had received surgery and [...] Read more.
This study aimed to investigate the performance of a deep learning-based survival-prediction model, which predicts the overall survival (OS) time of glioblastoma patients who have received surgery followed by concurrent chemoradiotherapy (CCRT). The medical records of glioblastoma patients who had received surgery and CCRT between January 2011 and December 2017 were retrospectively reviewed. Based on our inclusion criteria, 118 patients were selected and semi-randomly allocated to training and test datasets (3:1 ratio, respectively). A convolutional neural network–based deep learning model was trained with magnetic resonance imaging (MRI) data and clinical profiles to predict OS. The MRI was reconstructed by using four pulse sequences (22 slices) and nine images were selected based on the longest slice of glioblastoma by a physician for each pulse sequence. The clinical profiles consist of personal, genetic, and treatment factors. The concordance index (C-index) and integrated area under the curve (iAUC) of the time-dependent area-under-the-curve curves of each model were calculated to evaluate the performance of the survival-prediction models. The model that incorporated clinical and radiomic features showed a higher C-index (0.768 (95% confidence interval (CI): 0.759, 0.776)) and iAUC (0.790 (95% CI: 0.783, 0.797)) than the model using clinical features alone (C-index = 0.693 (95% CI: 0.685, 0.701); iAUC = 0.723 (95% CI: 0.716, 0.731)) and the model using radiomic features alone (C-index = 0.590 (95% CI: 0.579, 0.600); iAUC = 0.614 (95% CI: 0.607, 0.621)). These improvements to the C-indexes and iAUCs were validated using the 1000-times bootstrapping method; all were statistically significant (p < 0.001). This study suggests the synergistic benefits of using both clinical and radiomic parameters. Furthermore, it indicates the potential of multi-parametric deep learning models for the survival prediction of glioblastoma patients. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

14 pages, 4361 KiB  
Article
Combination of Peri-Tumoral and Intra-Tumoral Radiomic Features on Bi-Parametric MRI Accurately Stratifies Prostate Cancer Risk: A Multi-Site Study
by Ahmad Algohary, Rakesh Shiradkar, Shivani Pahwa, Andrei Purysko, Sadhna Verma, Daniel Moses, Ronald Shnier, Anne-Maree Haynes, Warick Delprado, James Thompson, Sreeharsha Tirumani, Amr Mahran, Ardeshir R Rastinehad, Lee Ponsky, Phillip D. Stricker and Anant Madabhushi
Cancers 2020, 12(8), 2200; https://doi.org/10.3390/cancers12082200 - 06 Aug 2020
Cited by 47 | Viewed by 4693
Abstract
Background: Prostate cancer (PCa) influences its surrounding habitat, which tends to manifest as different phenotypic appearances on magnetic resonance imaging (MRI). This region surrounding the PCa lesion, or the peri-tumoral region, may encode useful information that can complement intra-tumoral information to enable better [...] Read more.
Background: Prostate cancer (PCa) influences its surrounding habitat, which tends to manifest as different phenotypic appearances on magnetic resonance imaging (MRI). This region surrounding the PCa lesion, or the peri-tumoral region, may encode useful information that can complement intra-tumoral information to enable better risk stratification. Purpose: To evaluate the role of peri-tumoral radiomic features on bi-parametric MRI (T2-weighted and Diffusion-weighted) to distinguish PCa risk categories as defined by D’Amico Risk Classification System. Materials and Methods: We studied a retrospective, HIPAA-compliant, 4-institution cohort of 231 PCa patients (n = 301 lesions) who underwent 3T multi-parametric MRI prior to biopsy. PCa regions of interest (ROIs) were delineated on MRI by experienced radiologists following which peri-tumoral ROIs were defined. Radiomic features were extracted within the intra- and peri-tumoral ROIs. Radiomic features differentiating low-risk from: (1) high-risk (L-vs.-H), and (2) (intermediate- and high-risk (L-vs.-I + H)) lesions were identified. Using a multi-institutional training cohort of 151 lesions (D1, N = 116 patients), machine learning classifiers were trained using peri- and intra-tumoral features individually and in combination. The remaining 150 lesions (D2, N = 115 patients) were used for independent hold-out validation and were evaluated using Receiver Operating Characteristic (ROC) analysis and compared with PI-RADS v2 scores. Results: Validation on D2 using peri-tumoral radiomics alone resulted in areas under the ROC curve (AUCs) of 0.84 and 0.73 for the L-vs.-H and L-vs.-I + H classifications, respectively. The best combination of intra- and peri-tumoral features resulted in AUCs of 0.87 and 0.75 for the L-vs.-H and L-vs.-I + H classifications, respectively. This combination improved the risk stratification results by 3–6% compared to intra-tumoral features alone. Our radiomics-based model resulted in a 53% accuracy in differentiating L-vs.-H compared to PI-RADS v2 (48%), on the validation set. Conclusion: Our findings suggest that peri-tumoral radiomic features derived from prostate bi-parametric MRI add independent predictive value to intra-tumoral radiomic features for PCa risk assessment. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

20 pages, 2824 KiB  
Article
Radiomic Texture and Shape Descriptors of the Rectal Environment on Post-Chemoradiation T2-Weighted MRI are Associated with Pathologic Tumor Stage Regression in Rectal Cancers: A Retrospective, Multi-Institution Study
by Charlems Alvarez-Jimenez, Jacob T. Antunes, Nitya Talasila, Kaustav Bera, Justin T. Brady, Jayakrishna Gollamudi, Eric Marderstein, Matthew F. Kalady, Andrei Purysko, Joseph E. Willis, Sharon Stein, Kenneth Friedman, Rajmohan Paspulati, Conor P. Delaney, Eduardo Romero, Anant Madabhushi and Satish E. Viswanath
Cancers 2020, 12(8), 2027; https://doi.org/10.3390/cancers12082027 - 24 Jul 2020
Cited by 24 | Viewed by 4403
Abstract
(1) Background: The relatively poor expert restaging accuracy of MRI in rectal cancer after neoadjuvant chemoradiation may be due to the difficulties in visual assessment of residual tumor on post-treatment MRI. In order to capture underlying tissue alterations and morphologic changes in [...] Read more.
(1) Background: The relatively poor expert restaging accuracy of MRI in rectal cancer after neoadjuvant chemoradiation may be due to the difficulties in visual assessment of residual tumor on post-treatment MRI. In order to capture underlying tissue alterations and morphologic changes in rectal structures occurring due to the treatment, we hypothesized that radiomics texture and shape descriptors of the rectal environment (e.g., wall, lumen) on post-chemoradiation T2-weighted (T2w) MRI may be associated with tumor regression after neoadjuvant chemoradiation therapy (nCRT). (2) Methods: A total of 94 rectal cancer patients were retrospectively identified from three collaborating institutions, for whom a 1.5 or 3T T2w MRI was available after nCRT and prior to surgical resection. The rectal wall and the lumen were annotated by an expert radiologist on all MRIs, based on which 191 texture descriptors and 198 shape descriptors were extracted for each patient. (3) Results: Top-ranked features associated with pathologic tumor-stage regression were identified via cross-validation on a discovery set (n = 52, 1 institution) and evaluated via discriminant analysis in hold-out validation (n = 42, 2 institutions). The best performing features for distinguishing low (ypT0-2) and high (ypT3–4) pathologic tumor stages after nCRT comprised directional gradient texture expression and morphologic shape differences in the entire rectal wall and lumen. Not only were these radiomic features found to be resilient to variations in magnetic field strength and expert segmentations, a quadratic discriminant model combining them yielded consistent performance across multiple institutions (hold-out AUC of 0.73). (4) Conclusions: Radiomic texture and shape descriptors of the rectal wall from post-treatment T2w MRIs may be associated with low and high pathologic tumor stage after neoadjuvant chemoradiation therapy and generalized across variations between scanners and institutions. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Graphical abstract

18 pages, 2446 KiB  
Article
Pre-Treatment T2-WI Based Radiomics Features for Prediction of Locally Advanced Rectal Cancer Non-Response to Neoadjuvant Chemoradiotherapy: A Preliminary Study
by Bianca Petresc, Andrei Lebovici, Cosmin Caraiani, Diana Sorina Feier, Florin Graur and Mircea Marian Buruian
Cancers 2020, 12(7), 1894; https://doi.org/10.3390/cancers12071894 - 14 Jul 2020
Cited by 38 | Viewed by 3351
Abstract
Locally advanced rectal cancer (LARC) response to neoadjuvant chemoradiotherapy (nCRT) is very heterogeneous and up to 30% of patients are considered non-responders, presenting no tumor regression after nCRT. This study aimed to determine the ability of pre-treatment T2-weighted based radiomics features to predict [...] Read more.
Locally advanced rectal cancer (LARC) response to neoadjuvant chemoradiotherapy (nCRT) is very heterogeneous and up to 30% of patients are considered non-responders, presenting no tumor regression after nCRT. This study aimed to determine the ability of pre-treatment T2-weighted based radiomics features to predict LARC non-responders. A total of 67 LARC patients who underwent a pre-treatment MRI followed by nCRT and total mesorectal excision were assigned into training (n = 44) and validation (n = 23) groups. In both datasets, the patients were categorized according to the Ryan tumor regression grade (TRG) system into non-responders (TRG = 3) and responders (TRG 1 and 2). We extracted 960 radiomic features/patient from pre-treatment T2-weighted images. After a three-step feature selection process, including LASSO regression analysis, we built a radiomics score with seven radiomics features. This score was significantly higher among non-responders in both training and validation sets (p < 0.001 and p = 0.03) and it showed good predictive performance for LARC non-response, achieving an area under the curve (AUC) = 0.94 (95% CI: 0.82–0.99) in the training set and AUC = 0.80 (95% CI: 0.58–0.94) in the validation group. The multivariate analysis identified the radiomics score as an independent predictor for the tumor non-response (OR = 6.52, 95% CI: 1.87–22.72). Our results indicate that MRI radiomics features could be considered as potential imaging biomarkers for early prediction of LARC non-response to neoadjuvant treatment. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

16 pages, 2424 KiB  
Article
Potential Added Value of PET/CT Radiomics for Survival Prognostication beyond AJCC 8th Edition Staging in Oropharyngeal Squamous Cell Carcinoma
by Stefan P. Haider, Tal Zeevi, Philipp Baumeister, Christoph Reichel, Kariem Sharaf, Reza Forghani, Benjamin H. Kann, Benjamin L. Judson, Manju L. Prasad, Barbara Burtness, Amit Mahajan and Seyedmehdi Payabvash
Cancers 2020, 12(7), 1778; https://doi.org/10.3390/cancers12071778 - 03 Jul 2020
Cited by 35 | Viewed by 4345
Abstract
Accurate risk-stratification can facilitate precision therapy in oropharyngeal squamous cell carcinoma (OPSCC). We explored the potential added value of baseline positron emission tomography (PET)/computed tomography (CT) radiomic features for prognostication and risk stratification of OPSCC beyond the American Joint Committee on Cancer (AJCC) [...] Read more.
Accurate risk-stratification can facilitate precision therapy in oropharyngeal squamous cell carcinoma (OPSCC). We explored the potential added value of baseline positron emission tomography (PET)/computed tomography (CT) radiomic features for prognostication and risk stratification of OPSCC beyond the American Joint Committee on Cancer (AJCC) 8th edition staging scheme. Using institutional and publicly available datasets, we included OPSCC patients with known human papillomavirus (HPV) status, without baseline distant metastasis and treated with curative intent. We extracted 1037 PET and 1037 CT radiomic features quantifying lesion shape, imaging intensity, and texture patterns from primary tumors and metastatic cervical lymph nodes. Utilizing random forest algorithms, we devised novel machine-learning models for OPSCC progression-free survival (PFS) and overall survival (OS) using “radiomics” features, “AJCC” variables, and the “combined” set as input. We designed both single- (PET or CT) and combined-modality (PET/CT) models. Harrell’s C-index quantified survival model performance; risk stratification was evaluated in Kaplan–Meier analysis. A total of 311 patients were included. In HPV-associated OPSCC, the best “radiomics” model achieved an average C-index ± standard deviation of 0.62 ± 0.05 (p = 0.02) for PFS prediction, compared to 0.54 ± 0.06 (p = 0.32) utilizing “AJCC” variables. Radiomics-based risk-stratification of HPV-associated OPSCC was significant for PFS and OS. Similar trends were observed in HPV-negative OPSCC. In conclusion, radiomics imaging features extracted from pre-treatment PET/CT may provide complimentary information to the current AJCC staging scheme for survival prognostication and risk-stratification of HPV-associated OPSCC. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

14 pages, 1875 KiB  
Article
Multiparametric MRI for Prostate Cancer Characterization: Combined Use of Radiomics Model with PI-RADS and Clinical Parameters
by Piotr Woźnicki, Niklas Westhoff, Thomas Huber, Philipp Riffel, Matthias F. Froelich, Eva Gresser, Jost von Hardenberg, Alexander Mühlberg, Maurice Stephan Michel, Stefan O. Schoenberg and Dominik Nörenberg
Cancers 2020, 12(7), 1767; https://doi.org/10.3390/cancers12071767 - 02 Jul 2020
Cited by 73 | Viewed by 5054
Abstract
Radiomics is an emerging field of image analysis with potential applications in patient risk stratification. This study developed and evaluated machine learning models using quantitative radiomic features extracted from multiparametric magnetic resonance imaging (mpMRI) to detect and classify prostate cancer (PCa). In total, [...] Read more.
Radiomics is an emerging field of image analysis with potential applications in patient risk stratification. This study developed and evaluated machine learning models using quantitative radiomic features extracted from multiparametric magnetic resonance imaging (mpMRI) to detect and classify prostate cancer (PCa). In total, 191 patients that underwent prostatic mpMRI and combined targeted and systematic fusion biopsy were retrospectively included. Segmentations of the whole prostate glands and index lesions were performed manually in apparent diffusion coefficient (ADC) maps and T2-weighted MRI. Radiomic features were extracted from regions corresponding to the whole prostate gland and index lesion. The best performing combination of feature setup and classifier was selected to compare its predictive ability of the radiologist’s evaluation (PI-RADS), mean ADC, prostate specific antigen density (PSAD) and digital rectal examination (DRE) using receiver operating characteristic (ROC) analysis. Models were evaluated using repeated 5-fold cross-validation and a separate independent test cohort. In the test cohort, an ensemble model combining a radiomics model, with models for PI-RADS, PSAD and DRE achieved high predictive AUCs for the differentiation of (i) malignant from benign prostatic lesions (AUC = 0.889) and of (ii) clinically significant (csPCa) from clinically insignificant PCa (cisPCa) (AUC = 0.844). Our combined model was numerically superior to PI-RADS for cancer detection (AUC = 0.779; p = 0.054) as well as for clinical significance prediction (AUC = 0.688; p = 0.209) and showed a significantly better performance compared to mADC for csPCa prediction (AUC = 0.571; p = 0.022). In our study, radiomics accurately characterizes prostatic index lesions and shows performance comparable to radiologists for PCa characterization. Quantitative image data represent a potential biomarker, which, when combined with PI-RADS, PSAD and DRE, predicts csPCa more accurately than mADC. Prognostic machine learning models could assist in csPCa detection and patient selection for MRI-guided biopsy. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

20 pages, 3334 KiB  
Article
Multiparametric Analysis of Longitudinal Quantitative MRI Data to Identify Distinct Tumor Habitats in Preclinical Models of Breast Cancer
by Anum K. Syed, Jennifer G. Whisenant, Stephanie L. Barnes, Anna G. Sorace and Thomas E. Yankeelov
Cancers 2020, 12(6), 1682; https://doi.org/10.3390/cancers12061682 - 24 Jun 2020
Cited by 27 | Viewed by 4278
Abstract
This study identifies physiological tumor habitats from quantitative magnetic resonance imaging (MRI) data and evaluates their alterations in response to therapy. Two models of breast cancer (BT-474 and MDA-MB-231) were imaged longitudinally with diffusion-weighted MRI and dynamic contrast-enhanced MRI to quantify tumor cellularity [...] Read more.
This study identifies physiological tumor habitats from quantitative magnetic resonance imaging (MRI) data and evaluates their alterations in response to therapy. Two models of breast cancer (BT-474 and MDA-MB-231) were imaged longitudinally with diffusion-weighted MRI and dynamic contrast-enhanced MRI to quantify tumor cellularity and vascularity, respectively, during treatment with trastuzumab or albumin-bound paclitaxel. Tumors were stained for anti-CD31, anti-Ki-67, and H&E. Imaging and histology data were clustered to identify tumor habitats and percent tumor volume (MRI) or area (histology) of each habitat was quantified. Histological habitats were correlated with MRI habitats. Clustering of both the MRI and histology data yielded three clusters: high-vascularity high-cellularity (HV-HC), low-vascularity high-cellularity (LV-HC), and low-vascularity low-cellularity (LV-LC). At day 4, BT-474 tumors treated with trastuzumab showed a decrease in LV-HC (p = 0.03) and increase in HV-HC (p = 0.03) percent tumor volume compared to control. MDA-MB-231 tumors treated with low-dose albumin-bound paclitaxel showed a longitudinal decrease in LV-HC percent tumor volume at day 3 (p = 0.01). Positive correlations were found between histological and imaging-derived habitats: HV-HC (BT-474: p = 0.03), LV-HC (MDA-MB-231: p = 0.04), LV-LC (BT-474: p = 0.04; MDA-MB-231: p < 0.01). Physiologically distinct tumor habitats associated with therapeutic response were identified with MRI and histology data in preclinical models of breast cancer. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

16 pages, 2582 KiB  
Article
Association of a CT-Based Clinical and Radiomics Score of Non-Small Cell Lung Cancer (NSCLC) with Lymph Node Status and Overall Survival
by Francesca Botta, Sara Raimondi, Lisa Rinaldi, Federica Bellerba, Federica Corso, Vincenzo Bagnardi, Daniela Origgi, Rocco Minelli, Giovanna Pitoni, Francesco Petrella, Lorenzo Spaggiari, Alessio G. Morganti, Filippo Del Grande, Massimo Bellomi and Stefania Rizzo
Cancers 2020, 12(6), 1432; https://doi.org/10.3390/cancers12061432 - 31 May 2020
Cited by 34 | Viewed by 5916
Abstract
Background: To evaluate whether a model based on radiomic and clinical features may be associated with lymph node (LN) status and overall survival (OS) in lung cancer (LC) patients; to evaluate whether CT reconstruction algorithms may influence the model performance. Methods: patients operated [...] Read more.
Background: To evaluate whether a model based on radiomic and clinical features may be associated with lymph node (LN) status and overall survival (OS) in lung cancer (LC) patients; to evaluate whether CT reconstruction algorithms may influence the model performance. Methods: patients operated on for LC with a pathological stage up to T3N1 were retrospectively selected and divided into training and validation sets. For the prediction of positive LNs and OS, the Least Absolute Shrinkage and Selection Operator (LASSO) logistic regression model was used; univariable and multivariable logistic regression analysis assessed the association of clinical-radiomic variables and endpoints. All tests were repeated after dividing the groups according to the CT reconstruction algorithm. p-values < 0.05 were considered significant. Results: 270 patients were included and divided into training (n = 180) and validation sets (n = 90). Transfissural extension was significantly associated with positive LNs. For OS prediction, high- and low-risk groups were different according to the radiomics score, also after dividing the two groups according to reconstruction algorithms. Conclusions: a combined clinical–radiomics model was not superior to a single clinical or single radiomics model to predict positive LNs. A radiomics model was able to separate high-risk and low-risk patients for OS; CTs reconstructed with Iterative Reconstructions (IR) algorithm showed the best model performance. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

13 pages, 1446 KiB  
Article
[18F]FDG-PET/CT Radiomics for Prediction of Bone Marrow Involvement in Mantle Cell Lymphoma: A Retrospective Study in 97 Patients
by Marius E. Mayerhoefer, Christopher C. Riedl, Anita Kumar, Ahmet Dogan, Peter Gibbs, Michael Weber, Philipp B. Staber, Sandra Huicochea Castellanos and Heiko Schöder
Cancers 2020, 12(5), 1138; https://doi.org/10.3390/cancers12051138 - 02 May 2020
Cited by 22 | Viewed by 2760
Abstract
Biopsy is the standard for assessment of bone marrow involvement in mantle cell lymphoma (MCL). We investigated whether [18F]FDG-PET radiomic texture features can improve prediction of bone marrow involvement in MCL, compared to standardized uptake values (SUV), and whether combination with laboratory data [...] Read more.
Biopsy is the standard for assessment of bone marrow involvement in mantle cell lymphoma (MCL). We investigated whether [18F]FDG-PET radiomic texture features can improve prediction of bone marrow involvement in MCL, compared to standardized uptake values (SUV), and whether combination with laboratory data improves results. Ninety-seven MCL patients were retrospectively included. SUVmax, SUVmean, SUVpeak and 16 co-occurrence matrix texture features were extracted from pelvic bones on [18F]FDG-PET/CT. A multi-layer perceptron neural network was used to compare three combinations for prediction of bone marrow involvement—the SUVs, a radiomic signature based on SUVs and texture features, and the radiomic signature combined with laboratory parameters. This step was repeated using two cut-off values for relative bone marrow involvement: REL > 5% (>5% of red/cellular bone marrow); and REL > 10%. Biopsy demonstrated bone marrow involvement in 67/97 patients (69.1%). SUVs, the radiomic signature, and the radiomic signature with laboratory data showed AUCs of up to 0.66, 0.73, and 0.81 for involved vs. uninvolved bone marrow; 0.68, 0.84, and 0.84 for REL ≤ 5% vs. REL > 5%; and 0.69, 0.85, and 0.87 for REL ≤ 10% vs. REL > 10%. In conclusion, [18F]FDG-PET texture features improve SUV-based prediction of bone marrow involvement in MCL. The results may be further improved by combination with laboratory parameters. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

12 pages, 2839 KiB  
Article
Radiomics-Based Outcome Prediction for Pancreatic Cancer Following Stereotactic Body Radiotherapy
by Elsa Parr, Qian Du, Chi Zhang, Chi Lin, Ahsan Kamal, Josiah McAlister, Xiaoying Liang, Kyle Bavitz, Gerard Rux, Michael Hollingsworth, Michael Baine and Dandan Zheng
Cancers 2020, 12(4), 1051; https://doi.org/10.3390/cancers12041051 - 24 Apr 2020
Cited by 39 | Viewed by 4068
Abstract
(1) Background: Radiomics use high-throughput mining of medical imaging data to extract unique information and predict tumor behavior. Currently available clinical prediction models poorly predict treatment outcomes in pancreatic adenocarcinoma. Therefore, we used radiomic features of primary pancreatic tumors to develop outcome prediction [...] Read more.
(1) Background: Radiomics use high-throughput mining of medical imaging data to extract unique information and predict tumor behavior. Currently available clinical prediction models poorly predict treatment outcomes in pancreatic adenocarcinoma. Therefore, we used radiomic features of primary pancreatic tumors to develop outcome prediction models and compared them to traditional clinical models. (2) Methods: We extracted and analyzed radiomic data from pre-radiation contrast-enhanced CTs of 74 pancreatic cancer patients undergoing stereotactic body radiotherapy. A panel of over 800 radiomic features was screened to create overall survival and local-regional recurrence prediction models, which were compared to clinical prediction models and models combining radiomic and clinical information. (3) Results: A 6-feature radiomic signature was identified that achieved better overall survival prediction performance than the clinical model (mean concordance index: 0.66 vs. 0.54 on resampled cross-validation test sets), and the combined model improved the performance slightly further to 0.68. Similarly, a 7-feature radiomic signature better predicted recurrence than the clinical model (mean AUC of 0.78 vs. 0.66). (4) Conclusion: Overall survival and recurrence can be better predicted with models based on radiomic features than with those based on clinical features for pancreatic cancer. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

15 pages, 2483 KiB  
Article
External Validation of an MRI-Derived Radiomics Model to Predict Biochemical Recurrence after Surgery for High-Risk Prostate Cancer
by Vincent Bourbonne, Georges Fournier, Martin Vallières, François Lucia, Laurent Doucet, Valentin Tissot, Gilles Cuvelier, Stephane Hue, Henri Le Penn Du, Luc Perdriel, Nicolas Bertrand, Frederic Staroz, Dimitris Visvikis, Olivier Pradier, Mathieu Hatt and Ulrike Schick
Cancers 2020, 12(4), 814; https://doi.org/10.3390/cancers12040814 - 28 Mar 2020
Cited by 49 | Viewed by 3463
Abstract
Adjuvant radiotherapy after prostatectomy was recently challenged by early salvage radiotherapy, which highlighted the need for biomarkers to improve risk stratification. Therefore, we developed an MRI ADC map-derived radiomics model to predict biochemical recurrence (BCR) and BCR-free survival (bRFS) after surgery. Our goal [...] Read more.
Adjuvant radiotherapy after prostatectomy was recently challenged by early salvage radiotherapy, which highlighted the need for biomarkers to improve risk stratification. Therefore, we developed an MRI ADC map-derived radiomics model to predict biochemical recurrence (BCR) and BCR-free survival (bRFS) after surgery. Our goal in this work was to externally validate this radiomics-based prediction model. Experimental Design: A total of 195 patients with a high recurrence risk of prostate cancer (pT3-4 and/or R1 and/or Gleason’s score > 7) were retrospectively included in two institutions. Patients with postoperative PSA (Prostate Specific Antigen) > 0.04 ng/mL or lymph node involvement were excluded. Radiomics features were extracted from T2 and ADC delineated tumors. A total of 107 patients from Institution 1 were used to retrain the previously published model. The retrained model was then applied to 88 patients from Institution 2 for external validation. BCR predictions were evaluated using AUC (Area Under the Curve), accuracy, and bRFS using Kaplan–Meier curves. Results: With a median follow-up of 46.3 months, 52/195 patients experienced BCR. In the retraining cohort, the clinical prediction model (combining the number of risk factors and postoperative PSA) demonstrated moderate predictive power (accuracy of 63%). The radiomics model (ADC-based SZEGLSZM) predicted BCR with an accuracy of 78% and allowed for significant stratification of patients for bRFS (p < 0.0001). In Institution 2, this radiomics model remained predictive of BCR (accuracy of 0.76%) contrary to the clinical model (accuracy of 0.56%). Conclusions: The recently developed MRI ADC map-based radiomics model was validated in terms of its predictive accuracy of BCR and bRFS after prostatectomy in an external cohort. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

11 pages, 2305 KiB  
Article
Extended Texture Analysis of Non-Enhanced Whole-Body MRI Image Data for Response Assessment in Multiple Myeloma Patients Undergoing Systemic Therapy
by Kaspar Ekert, Clemens Hinterleitner, Karolin Baumgartner, Jan Fritz and Marius Horger
Cancers 2020, 12(3), 761; https://doi.org/10.3390/cancers12030761 - 24 Mar 2020
Cited by 38 | Viewed by 3488
Abstract
Identifying MRI-based radiomics features capable to assess response to systemic treatment in multiple myeloma (MM) patients. Retrospective analysis of whole-body MR-image data in 67 consecutive stage III MM patients (40 men; mean age, 60.4 years). Bone marrow involvement was evaluated using a standardized [...] Read more.
Identifying MRI-based radiomics features capable to assess response to systemic treatment in multiple myeloma (MM) patients. Retrospective analysis of whole-body MR-image data in 67 consecutive stage III MM patients (40 men; mean age, 60.4 years). Bone marrow involvement was evaluated using a standardized MR-imaging protocol consisting of T1w-, short-tau inversion recovery- (STIR-) and diffusion-weighted-imaging (DWI) sequences. Ninety-two radiomics features were evaluated, both in focally and diffusely involved bone marrow. Volumes of interest (VOI) were used. Response to treatment was classified according to International Myeloma Working Group (IMWG) criteria in complete response (CR), very-good and/or partial response (VGPR + PR), and non-response (stable disease (SD) and progressive disease (PD)). According to the IMWG-criteria, response categories were CR (n = 35), VGPR + PR (n = 19), and non-responders (n = 13). On apparent diffusion coefficient (ADC)-maps, gray-level small size matrix small area emphasis (Gray Level Size Zone (GLSZM) small area emphasis (SAE)) significantly correlated with CR (p < 0.001), whereas GLSZM non-uniformity normalized (NUN) significantly (p < 0.008) with VGPR/PR in focal medullary lesions (FL), whereas in diffuse involvement, 1st order root mean squared significantly (p < 0.001) correlated with CR, whereas for VGPR/PR Log (gray-level run-length matrix (GLRLM) Short Run High Gray Level Emphasis) proved significant (p < 0.003). On T1w, GLRLM NUN significantly (p < 0.002) correlated with CR in FL, whereas gray-level co-occurrence matric (GLCM) informational measure of correlation (Imc1) significantly (p < 0.04) correlated with VGPR/PR. For diffuse myeloma involvement, neighboring gray-tone difference matrix (NGTDM) contrast and 1st order skewness were significantly associated with CR and VGPR/PR (p < 0.001 for both). On STIR-images, CR correlated with gray-level co-occurrence matrix (GLCM) Informational Measure of Correlation (IMC) 1 (p < 0.001) in FL and 1st order mean absolute deviation in diffusely involved bone marrow (p < 0.001). VGPR/PR correlated at best in FL with GSZLM size zone NUN (p < 0.019) and in all other involved medullary areas with GLSZM large area low gray level emphasis (p < 0.001). GLSZM large area low gray level emphasis also significantly correlated with the degree of bone marrow infiltration assessed histologically (p = 0.006). GLCM IMC 1 proved significant throughout T1w/STIR sequences, whereas GLSZM NUN in STIR and ADC. MRI-based texture features proved significant to assess clinical and hematological response (CR, VPGR, and PR) in multiple myeloma patients undergoing systemic treatment. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

9 pages, 1343 KiB  
Article
Radiomics Model Based on Non-Contrast CT Shows No Predictive Power for Complete Pathological Response in Locally Advanced Rectal Cancer
by Gordian Hamerla, Hans-Jonas Meyer, Peter Hambsch, Ulrich Wolf, Thomas Kuhnt, Karl-Titus Hoffmann and Alexey Surov
Cancers 2019, 11(11), 1680; https://doi.org/10.3390/cancers11111680 - 29 Oct 2019
Cited by 23 | Viewed by 2761
Abstract
(1) Background: About 15% of the patients undergoing neoadjuvant chemoradiation for locally advanced rectal cancer exhibit pathological complete response (pCR). The surgical approach is associated with major risks as well as a potential negative impact on quality of life and has been questioned [...] Read more.
(1) Background: About 15% of the patients undergoing neoadjuvant chemoradiation for locally advanced rectal cancer exhibit pathological complete response (pCR). The surgical approach is associated with major risks as well as a potential negative impact on quality of life and has been questioned in the past. Still, there is no evidence of a reliable clinical or radiological surrogate marker for pCR. This study aims to replicate previously reported response predictions on the basis of non-contrast CT scans on an independent patient cohort. (2) Methods: A total of 169 consecutive patients (126 males, 43 females) that underwent neoadjuvant chemoradiation and consecutive total mesorectal excision were included. The solid tumors were segmented on CT scans acquired on the same scanner for treatment planning. To quantify intratumoral 3D spatial heterogeneity, 1819 radiomics parameters were derived per case. Feature selection and algorithmic modeling were performed to classify pCR vs. non-pCR cases. A random forest model was trained on the dataset using 4-fold cross-validation. (3) Results: The model achieved an accuracy of 87%, higher than previously reported. Correction for the imbalanced distribution of pCR and non-PCR cases (13% and 87% respectively) was applied, yielding a balanced accuracy score of 0.5%. An additional experiment to classify a computer-generated random data sample using the same model led to comparable results. (4) Conclusions: There is no evidence of added value of a radiomics model based on on-contrast CT scans for prediction of pCR in rectal cancer. The imbalance of the target variable could be identified as a key issue, leading to a biased model and optimistic predictions. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

Review

Jump to: Research, Other

13 pages, 1614 KiB  
Review
Automated Classification of Significant Prostate Cancer on MRI: A Systematic Review on the Performance of Machine Learning Applications
by Jose M. Castillo T., Muhammad Arif, Wiro J. Niessen, Ivo G. Schoots and Jifke F. Veenland
Cancers 2020, 12(6), 1606; https://doi.org/10.3390/cancers12061606 - 17 Jun 2020
Cited by 52 | Viewed by 4821
Abstract
Significant prostate carcinoma (sPCa) classification based on MRI using radiomics or deep learning approaches has gained much interest, due to the potential application in assisting in clinical decision-making. Objective: To systematically review the literature (i) to determine which algorithms are most frequently used [...] Read more.
Significant prostate carcinoma (sPCa) classification based on MRI using radiomics or deep learning approaches has gained much interest, due to the potential application in assisting in clinical decision-making. Objective: To systematically review the literature (i) to determine which algorithms are most frequently used for sPCa classification, (ii) to investigate whether there exists a relation between the performance and the method or the MRI sequences used, (iii) to assess what study design factors affect the performance on sPCa classification, and (iv) to research whether performance had been evaluated in a clinical setting Methods: The databases Embase and Ovid MEDLINE were searched for studies describing machine learning or deep learning classification methods discriminating between significant and nonsignificant PCa on multiparametric MRI that performed a valid validation procedure. Quality was assessed by the modified radiomics quality score. We computed the median area under the receiver operating curve (AUC) from overall methods and the interquartile range. Results: From 2846 potentially relevant publications, 27 were included. The most frequent algorithms used in the literature for PCa classification are logistic regression (22%) and convolutional neural networks (CNNs) (22%). The median AUC was 0.79 (interquartile range: 0.77–0.87). No significant effect of number of included patients, image sequences, or reference standard on the reported performance was found. Three studies described an external validation and none of the papers described a validation in a prospective clinical trial. Conclusions: To unlock the promising potential of machine and deep learning approaches, validation studies and clinical prospective studies should be performed with an established protocol to assess the added value in decision-making. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

Other

Jump to: Research, Review

19 pages, 1378 KiB  
Perspective
Radiomics at a Glance: A Few Lessons Learned from Learning Approaches
by Enrico Capobianco and Jun Deng
Cancers 2020, 12(9), 2453; https://doi.org/10.3390/cancers12092453 - 29 Aug 2020
Cited by 7 | Viewed by 2816
Abstract
Processing and modeling medical images have traditionally represented complex tasks requiring multidisciplinary collaboration. The advent of radiomics has assigned a central role to quantitative data analytics targeting medical image features algorithmically extracted from large volumes of images. Apart from the ultimate goal of [...] Read more.
Processing and modeling medical images have traditionally represented complex tasks requiring multidisciplinary collaboration. The advent of radiomics has assigned a central role to quantitative data analytics targeting medical image features algorithmically extracted from large volumes of images. Apart from the ultimate goal of supporting diagnostic, prognostic, and therapeutic decisions, radiomics is computationally attractive due to specific strengths: scalability, efficiency, and precision. Optimization is achieved by highly sophisticated statistical and machine learning algorithms, but it is especially deep learning that stands out as the leading inference approach. Various types of hybrid learning can be considered when building complex integrative approaches aimed to deliver gains in accuracy for both classification and prediction tasks. This perspective reviews some selected learning methods by focusing on both their significance for radiomics and their unveiled potential. Full article
(This article belongs to the Special Issue Radiomics and Cancers)
Show Figures

Figure 1

Back to TopTop