1
|
Chirra PV, Giriprakash P, Rizk AG, Kurowski JA, Viswanath SE, Gandhi NS. Developing a Reproducible Radiomics Model for Diagnosis of Active Crohn's Disease on CT Enterography Across Annotation Variations and Acquisition Differences. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025; 38:1594-1605. [PMID: 39466507 DOI: 10.1007/s10278-024-01303-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024]
Abstract
To systematically identify radiomics features on CT enterography (CTE) scans which can accurately diagnose active Crohn's disease across multiple sources of variation. Retrospective study of CTE scans curated between 2013 and 2015, comprising 164 subjects (65 male, 99 female; all patients were over the age of 18) with endoscopic confirmation for the presence or absence of active Crohn's disease. All patients had three distinct sets of scans available (full and reduced dose, where the latter had been reconstructed via two different methods), acquired on a single scanner at a single institution. Radiomics descriptors from annotated terminal ileum regions were individually and systematically evaluated for resilience to different imaging variations (changes in dose/reconstruction, batch effects, and simulated annotation differences) via multiple reproducibility measures. Multiple radiomics models (by accounting for each source of variation) were evaluated in terms of classifier area under the ROC curve (AUC) for identifying patients with active Crohn's disease, across separate discovery and hold-out validation cohorts. Radiomics descriptors selected based on resiliency to multiple sources of imaging variation yielded the highest overall classification performance in the discovery cohort (AUC = 0.79 ± 0.04) which also best generalized in hold-out validation (AUC = 0.81). Performance was maintained across multiple doses and reconstructions while also being significantly better (p < 0.001) than non-resilient descriptors or descriptors only resilient to a single source of variation. Radiomics features can accurately diagnose active Crohn's disease on CTE scans across multiple sources of imaging variation via systematic analysis of reproducibility measures. Clinical utility and translatability of radiomics features for diagnosis and characterization of Crohn's disease on CTE scans will be contingent on their reproducibility across multiple types and sources of imaging variation.
Collapse
Affiliation(s)
- Prathyush V Chirra
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Pavithran Giriprakash
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Alain G Rizk
- Section, Abdominal Imaging, Imaging Institute, and Digestive Diseases and Surgery Institute and Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jacob A Kurowski
- Department of Pediatric Gastroenterology, Hepatology & Nutrition, Cleveland Clinic, Cleveland, OH, USA
| | - Satish E Viswanath
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
- Cleveland Veterns Affairs Medical Center, Cleveland, OH, USA.
| | - Namita S Gandhi
- Section, Abdominal Imaging, Imaging Institute, and Digestive Diseases and Surgery Institute and Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
2
|
Choi J, Gordon A, Eresen A, Zhang Z, Borhani A, Bagci U, Lewandowski R, Kim DH. Current applications of radiomics in the assessment of tumor microenvironment of hepatocellular carcinoma. Abdom Radiol (NY) 2025:10.1007/s00261-025-04916-w. [PMID: 40208284 DOI: 10.1007/s00261-025-04916-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/10/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025]
Abstract
The tumor microenvironment (TME) of hepatocellular carcinoma (HCC) has garnered significant attention, especially with the rise of immunotherapy as a treatment strategy. Radiomics, an innovative technique, offers valuable insights into the intricate structure of the TME. This review highlights recent advancements in radiomics for analyzing the HCC TME, identifies key areas that warrant further research, and explores comprehensive multi-omics approaches that extend the potential of radiomics to new frontiers.
Collapse
Affiliation(s)
- Junghwa Choi
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Andrew Gordon
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Aydin Eresen
- Department of Radiological Sciences, University of California, Irvine, Irvine, USA
| | - Zhuoli Zhang
- Department of Radiological Sciences, University of California, Irvine, Irvine, USA
| | - Amir Borhani
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Ulas Bagci
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Robert Lewandowski
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, 60611, USA.
| |
Collapse
|
3
|
Zhou C, Xin H, Qian L, Zhang Y, Wang J, Luo J. Computed Tomography-Based Habitat Analysis for Prognostic Stratification in Colorectal Liver Metastases. CANCER INNOVATION 2025; 4:e70000. [PMID: 40078361 PMCID: PMC11897531 DOI: 10.1002/cai2.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 12/05/2024] [Accepted: 12/19/2024] [Indexed: 03/14/2025]
Abstract
Background Colorectal liver metastasis (CRLM) has a poor prognosis, and traditional prognostic models have certain limitations in clinical application. This study aims to evaluate the prognostic value of CT-based habitat analysis in CRLM patients and compare it with existing traditional prognostic models to provide more evidence for individualized treatment of CRLM patients. Methods This retrospective study included 197 patients with CRLM whose preoperative contrast-enhanced CT images and corresponding DICOM Segmentation Objects (DSOs) were obtained from The Cancer Imaging Archive (TCIA). Tumor regions were segmented, and habitat features representing distinct subregions were extracted. An unsupervised K-means clustering algorithm classified the tumors into two clusters based on their habitat characteristics. Kaplan-Meier analysis was used to evaluate overall survival (OS), disease-free survival (DFS), and liver-specific DFS. The habitat model's predictive performance was compared with the Clinical Risk Score (CRS) and Tumor Burden Score (TBS) using the concordance index (C-index), Integrated Brier Score (IBS), and time-dependent area under the curve (AUC). Results The habitat model identified two distinct patient clusters with significant differences in OS, DFS, and liver-specific DFS (p < 0.01). Compared with CRS and TBS, the habitat model demonstrated superior predictive accuracy, particularly for DFS and liver-specific DFS, with higher time-dependent AUC values and improved model calibration (lower IBS). Conclusions CT-based habitat analysis captures spatial tumor heterogeneity and provides enhanced prognostic stratification in CRLM. The method outperforms conventional models and offers potential for more personalized treatment planning.
Collapse
Affiliation(s)
- Chaoqun Zhou
- Department of Pathology, Huaihe HospitalHenan UniversityKaifengHenanChina
| | - Hao Xin
- Department of Radiology, Huaihe HospitalHenan UniversityKaifengHenanChina
| | - Lihua Qian
- Department of Pathology, Huaihe HospitalHenan UniversityKaifengHenanChina
| | - Yong Zhang
- Department of Biological TherapyHenan Provincial Cancer HospitalZhengzhouHenanChina
| | - Jing Wang
- Department of General SurgeryFirst Medical Center of PLA General HospitalBeijingChina
| | - Junpeng Luo
- Translational Medical Center, Huaihe HospitalHenan UniversityKaifengHenanChina
| |
Collapse
|
4
|
Li R, Bing X, Su X, Zhang C, Sun H, Dai Z, Ouyang A. The potential value of dual-energy CT radiomics in evaluating CD8 +, CD163 + and αSMA + cells in the tumor microenvironment of clear cell renal cell carcinoma. Clin Transl Oncol 2025; 27:716-726. [PMID: 39083142 DOI: 10.1007/s12094-024-03637-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/19/2024] [Indexed: 02/01/2025]
Abstract
PURPOSE This study aims to develop radiomics models and a nomogram based on machine learning techniques, preoperative dual-energy computed tomography (DECT) images, clinical and pathological characteristics, to explore the tumor microenvironment (TME) of clear cell renal cell carcinoma (ccRCC). METHODS We retrospectively recruited of 87 patients diagnosed with ccRCC through pathological confirmation from Center I (training set, n = 69; validation set, n = 18), and collected their DECT images and clinical information. Feature selection was conducted using variance threshold, SelectKBest, and the least absolute shrinkage and selection operator (LASSO). Radiomics models were then established using 14 classifiers to predict TME cells. Subsequently, we selected the most predictive radiomics features to calculate the radiomics score (Radscore). A combined model was constructed through multivariate logistic regression analysis combining the Radscore and relevant clinical characteristics, and presented in the form of a nomogram. Additionally, 17 patients were recruited from Center II as an external validation cohort for the nomogram. The performance of the models was assessed using methods such as the area under the receiver operating characteristic curve (AUC), calibration curve, and decision curve analysis (DCA). RESULTS The validation set AUC values for the radiomics models assessing CD8+, CD163+, and αSMA+ cells were 0.875, 0.889, and 0.864, respectively. Additionally, the external validation cohort AUC value for the nomogram reaches 0.849 and shows good calibration. CONCLUSION Radiomics models could allow for non-invasive assessment of TME cells from DECT images in ccRCC patients, promising to enhance our understanding and management of the tumor.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/diagnostic imaging
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/immunology
- Kidney Neoplasms/diagnostic imaging
- Kidney Neoplasms/pathology
- Kidney Neoplasms/immunology
- Male
- Female
- Tumor Microenvironment/immunology
- Middle Aged
- Retrospective Studies
- Tomography, X-Ray Computed/methods
- Nomograms
- Aged
- CD163 Antigen
- Antigens, CD/analysis
- Antigens, Differentiation, Myelomonocytic/analysis
- Antigens, Differentiation, Myelomonocytic/metabolism
- Receptors, Cell Surface/analysis
- Adult
- Machine Learning
- Radiomics
Collapse
Affiliation(s)
- Ruobing Li
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, 105 JieFang Road, Jinan, 250013, China
- Shandong First Medical University, Jinan, 250117, China
| | - Xue Bing
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, 105 JieFang Road, Jinan, 250013, China
| | - Xinyou Su
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Chunling Zhang
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, 105 JieFang Road, Jinan, 250013, China
| | - Haitao Sun
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, 105 JieFang Road, Jinan, 250013, China
| | - Zhengjun Dai
- Scientific Research Department, Huiying Medical Technology Co, Ltd, Beijing, 100192, China
| | - Aimei Ouyang
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, 105 JieFang Road, Jinan, 250013, China.
| |
Collapse
|
5
|
Fajemisin JA, Bryant JM, Saghand PG, Mills MN, Latifi K, Moros EG, Feygelman V, Frakes JM, Hoffe SE, Mittauer KE, Kutuk T, Kotecha R, El Naqa I, Rosenberg SA. Delta-Radiomics Using Machine Learning Classifiers With Auxiliary Data Sets to Predict Disease Progression During Magnetic Resonance-Guided Radiotherapy in Adrenal Metastases. JCO Clin Cancer Inform 2025; 9:e2400002. [PMID: 39854670 DOI: 10.1200/cci.24.00002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 10/23/2024] [Accepted: 12/17/2024] [Indexed: 01/26/2025] Open
Abstract
PURPOSE Adaptive radiotherapy accounts for interfractional anatomic changes. We hypothesize that changes in the gross tumor volumes identified during daily scans could be analyzed using delta-radiomics to predict disease progression events. We evaluated whether an auxiliary data set could improve prediction performance. MATERIALS AND METHODS We analyzed 108 patients (n = 90 internal; n = 18 external) who received ablative radiotherapy. The internal data set included 42 patients with adrenal cancer, 23 patients with lung cancer, and 25 patients with pancreatic cancer, with the clinical end point of progression-free survival events. The median dose was 50 Gy, which was delivered over five fractions. The delta features are the ratio of the features of the last to first treatment fraction, F5/F1, and the concatenation of the first and last fraction features, F1||F5. Decision tree classifier with and without auxiliary data sets, and the external data set was used exclusively for independent testing of the final models. RESULTS During internal training, for the F1||F5 model, the inclusion of the lung data set increased our AUC receiver operator characteristic curve (ROC) from 0.53 ± 0.12 to 0.61 ± 0.11, whereas the pancreatic data set increased our AUC-ROC to 0.60 ± 0.14. For the F5/F1 model, the inclusion of the lung auxiliary data increased our AUC-ROC from 0.52 ± 0.13 to 0.65 ± 0.11, whereas it modestly changed by 0.62 ± 0.13 with the pancreas. During external testing, for the F5/F1 model, we reported an AUC-ROC of 0.60 with the lung auxiliary data and 0.43 with the pancreatic data. Also, for the F5||F1 model, we reported an AUC-ROC of 0.70 with the lung auxiliary and 0.60 with the pancreatic data. CONCLUSION Decision trees provided an explainable model on the external data set. The validation of our model on an external data set may be the first step to biologically adapted radiotherapy recognizing radiomics signals for potential recurrence.
Collapse
Affiliation(s)
- Jesutofunmi A Fajemisin
- Machine Learning Department, H. Lee Moffit Cancer Center and Research Institute, Tampa, FL
- Department of Physics, University of South Florida, Tampa, FL
| | - John M Bryant
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | - Matthew N Mills
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Kujtim Latifi
- Machine Learning Department, H. Lee Moffit Cancer Center and Research Institute, Tampa, FL
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Eduardo G Moros
- Machine Learning Department, H. Lee Moffit Cancer Center and Research Institute, Tampa, FL
- Department of Physics, University of South Florida, Tampa, FL
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Vladimir Feygelman
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Jessica M Frakes
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Sarah E Hoffe
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Kathryn E Mittauer
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| | - Tugce Kutuk
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| | - Rupesh Kotecha
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| | - Issam El Naqa
- Machine Learning Department, H. Lee Moffit Cancer Center and Research Institute, Tampa, FL
- Department of Physics, University of South Florida, Tampa, FL
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Stephen A Rosenberg
- Machine Learning Department, H. Lee Moffit Cancer Center and Research Institute, Tampa, FL
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
6
|
Tran K, Ginzburg D, Hong W, Attenberger U, Ko HS. Post-radiotherapy stage III/IV non-small cell lung cancer radiomics research: a systematic review and comparison of CLEAR and RQS frameworks. Eur Radiol 2024; 34:6527-6543. [PMID: 38625613 PMCID: PMC11399214 DOI: 10.1007/s00330-024-10736-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/07/2024] [Accepted: 03/04/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Lung cancer, the second most common cancer, presents persistently dismal prognoses. Radiomics, a promising field, aims to provide novel imaging biomarkers to improve outcomes. However, clinical translation faces reproducibility challenges, despite efforts to address them with quality scoring tools. OBJECTIVE This study had two objectives: 1) identify radiomics biomarkers in post-radiotherapy stage III/IV nonsmall cell lung cancer (NSCLC) patients, 2) evaluate research quality using the CLEAR (CheckList_for_EvaluAtion_of_Radiomics_research), RQS (Radiomics_Quality_Score) frameworks, and formulate an amalgamated CLEAR-RQS tool to enhance scientific rigor. MATERIALS AND METHODS A systematic literature review (Jun-Aug 2023, MEDLINE/PubMed/SCOPUS) was conducted concerning stage III/IV NSCLC, radiotherapy, and radiomic features (RF). Extracted data included study design particulars, such as sample size, radiotherapy/CT technique, selected RFs, and endpoints. CLEAR and RQS were merged into a CLEAR-RQS checklist. Three readers appraised articles utilizing CLEAR, RQS, and CLEAR-RQS metrics. RESULTS Out of 871 articles, 11 met the inclusion/exclusion criteria. The Median cohort size was 91 (range: 10-337) with 9 studies being single-center. No common RF were identified. The merged CLEAR-RQS checklist comprised 61 items. Most unreported items were within CLEAR's "methods" and "open-source," and within RQS's "phantom-calibration," "registry-enrolled prospective-trial-design," and "cost-effective-analysis" sections. No study scored above 50% on RQS. Median CLEAR scores were 55.74% (32.33/58 points), and for RQS, 17.59% (6.3/36 points). CLEAR-RQS article ranking fell between CLEAR and RQS and aligned with CLEAR. CONCLUSION Radiomics research in post-radiotherapy stage III/IV NSCLC exhibits variability and frequently low-quality reporting. The formulated CLEAR-RQS checklist may facilitate education and holds promise for enhancing radiomics research quality. CLINICAL RELEVANCE STATEMENT Current radiomics research in the field of stage III/IV postradiotherapy NSCLC is heterogenous, lacking reproducibility, with no identified imaging biomarker. Radiomics research quality assessment tools may enhance scientific rigor and thereby facilitate radiomics translation into clinical practice. KEY POINTS There is heterogenous and low radiomics research quality in postradiotherapy stage III/IV nonsmall cell lung cancer. Barriers to reproducibility are small cohort size, nonvalidated studies, missing technical parameters, and lack of data, code, and model sharing. CLEAR (CheckList_for_EvaluAtion_of_Radiomics_research), RQS (Radiomics_Quality_Score), and the amalgamated CLEAR-RQS tool are useful frameworks for assessing radiomics research quality and may provide a valuable resource for educational purposes in the field of radiomics.
Collapse
Affiliation(s)
- Kevin Tran
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville, VIC 3052, Australia
| | - Daniel Ginzburg
- Department of Diagnostic and Interventional Radiology, Venusberg Campus 1, 53127, Bonn, Germany
| | - Wei Hong
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Ulrike Attenberger
- Department of Diagnostic and Interventional Radiology, Venusberg Campus 1, 53127, Bonn, Germany
| | - Hyun Soo Ko
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
- Department of Diagnostic and Interventional Radiology, Venusberg Campus 1, 53127, Bonn, Germany.
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, 305 Grattan St, Melbourne, VIC 3000, Australia.
| |
Collapse
|
7
|
Huang YS, Chen JLY, Ko WC, Chang YH, Chang CH, Chang YC. Clinical Variables and Radiomics Features for Predicting Pneumothorax in Patients Undergoing CT-guided Transthoracic Core Needle Biopsy. Radiol Cardiothorac Imaging 2024; 6:e230278. [PMID: 38780426 PMCID: PMC11211933 DOI: 10.1148/ryct.230278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 02/02/2024] [Accepted: 03/27/2024] [Indexed: 05/25/2024]
Abstract
Purpose To develop a prediction model combining both clinical and CT texture analysis radiomics features for predicting pneumothorax complications in patients undergoing CT-guided core needle biopsy. Materials and Methods A total of 424 patients (mean age, 65.6 years ± 12.7 [SD]; 232 male, 192 female) who underwent CT-guided core needle biopsy between January 2021 and October 2022 were retrospectively included as the training data set. Clinical and procedure-related characteristics were documented. Texture analysis radiomics features were extracted from the subpleural lung parenchyma traversed by needle. Moderate pneumothorax was defined as a postprocedure air rim of 2 cm or greater. The prediction model was developed using logistic regression with backward elimination, presented by linear fusion of the selected features weighted by their coefficients. Model performance was assessed using the area under the receiver operating characteristic curve (AUC). Validation was conducted in an external cohort (n = 45; mean age, 58.2 years ± 12.7; 19 male, 26 female) from a different hospital. Results Moderate pneumothorax occurred in 12.0% (51 of 424) of the training cohort and 8.9% (four of 45) of the external test cohort. Patients with emphysema (P < .001) or a longer needle path length (P = .01) exhibited a higher incidence of moderate pneumothorax in the training cohort. Texture analysis features, including gray-level co-occurrence matrix cluster shade (P < .001), gray-level run-length matrix low gray-level run emphasis (P = .049), gray-level run-length matrix run entropy (P = .003), gray-level size-zone matrix gray-level variance (P < .001), and neighboring gray-tone difference matrix complexity (P < .001), showed higher values in patients with moderate pneumothorax. The combined clinical-radiomics model demonstrated satisfactory performance in both the training (AUC 0.78, accuracy = 71.9%) and external test cohorts (AUC 0.86, accuracy 73.3%). Conclusion The model integrating both clinical and radiomics features offered practical diagnostic performance and accuracy for predicting moderate pneumothorax in patients undergoing CT-guided core needle biopsy. Keywords: Biopsy/Needle Aspiration, Thorax, CT, Pneumothorax, Core Needle Biopsy, Texture Analysis, Radiomics, CT Supplemental material is available for this article. © RSNA, 2024.
Collapse
Affiliation(s)
- Yu-Sen Huang
- From the Department of Medical Imaging (Y.S.H., W.C.K., Y.C.C.) and
Statistical Consulting Unit (Y.H.C., C.H.C.), National Taiwan University
Hospital, No. 7 Chung-Shan S. Rd, Taipei 100, Taiwan; Department of Radiology,
National Taiwan University College of Medicine, Taipei, Taiwan (Y.S.H.,
J.L.Y.C., Y.C.C.); and Department of Radiation Oncology, National Taiwan
University Cancer Center, Taipei, Taiwan (J.L.Y.C.)
| | - Jenny Ling-Yu Chen
- From the Department of Medical Imaging (Y.S.H., W.C.K., Y.C.C.) and
Statistical Consulting Unit (Y.H.C., C.H.C.), National Taiwan University
Hospital, No. 7 Chung-Shan S. Rd, Taipei 100, Taiwan; Department of Radiology,
National Taiwan University College of Medicine, Taipei, Taiwan (Y.S.H.,
J.L.Y.C., Y.C.C.); and Department of Radiation Oncology, National Taiwan
University Cancer Center, Taipei, Taiwan (J.L.Y.C.)
| | - Wei-Chun Ko
- From the Department of Medical Imaging (Y.S.H., W.C.K., Y.C.C.) and
Statistical Consulting Unit (Y.H.C., C.H.C.), National Taiwan University
Hospital, No. 7 Chung-Shan S. Rd, Taipei 100, Taiwan; Department of Radiology,
National Taiwan University College of Medicine, Taipei, Taiwan (Y.S.H.,
J.L.Y.C., Y.C.C.); and Department of Radiation Oncology, National Taiwan
University Cancer Center, Taipei, Taiwan (J.L.Y.C.)
| | - Yu-Han Chang
- From the Department of Medical Imaging (Y.S.H., W.C.K., Y.C.C.) and
Statistical Consulting Unit (Y.H.C., C.H.C.), National Taiwan University
Hospital, No. 7 Chung-Shan S. Rd, Taipei 100, Taiwan; Department of Radiology,
National Taiwan University College of Medicine, Taipei, Taiwan (Y.S.H.,
J.L.Y.C., Y.C.C.); and Department of Radiation Oncology, National Taiwan
University Cancer Center, Taipei, Taiwan (J.L.Y.C.)
| | - Chin-Hao Chang
- From the Department of Medical Imaging (Y.S.H., W.C.K., Y.C.C.) and
Statistical Consulting Unit (Y.H.C., C.H.C.), National Taiwan University
Hospital, No. 7 Chung-Shan S. Rd, Taipei 100, Taiwan; Department of Radiology,
National Taiwan University College of Medicine, Taipei, Taiwan (Y.S.H.,
J.L.Y.C., Y.C.C.); and Department of Radiation Oncology, National Taiwan
University Cancer Center, Taipei, Taiwan (J.L.Y.C.)
| | - Yeun-Chung Chang
- From the Department of Medical Imaging (Y.S.H., W.C.K., Y.C.C.) and
Statistical Consulting Unit (Y.H.C., C.H.C.), National Taiwan University
Hospital, No. 7 Chung-Shan S. Rd, Taipei 100, Taiwan; Department of Radiology,
National Taiwan University College of Medicine, Taipei, Taiwan (Y.S.H.,
J.L.Y.C., Y.C.C.); and Department of Radiation Oncology, National Taiwan
University Cancer Center, Taipei, Taiwan (J.L.Y.C.)
| |
Collapse
|
8
|
Derbal Y. Adaptive Cancer Therapy in the Age of Generative Artificial Intelligence. Cancer Control 2024; 31:10732748241264704. [PMID: 38897721 PMCID: PMC11189021 DOI: 10.1177/10732748241264704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024] Open
Abstract
Therapeutic resistance is a major challenge facing the design of effective cancer treatments. Adaptive cancer therapy is in principle the most viable approach to manage cancer's adaptive dynamics through drug combinations with dose timing and modulation. However, there are numerous open issues facing the clinical success of adaptive therapy. Chief among these issues is the feasibility of real-time predictions of treatment response which represent a bedrock requirement of adaptive therapy. Generative artificial intelligence has the potential to learn prediction models of treatment response from clinical, molecular, and radiomics data about patients and their treatments. The article explores this potential through a proposed integration model of Generative Pre-Trained Transformers (GPTs) in a closed loop with adaptive treatments to predict the trajectories of disease progression. The conceptual model and the challenges facing its realization are discussed in the broader context of artificial intelligence integration in oncology.
Collapse
Affiliation(s)
- Youcef Derbal
- Ted Rogers School of Information Technology Management, Toronto Metropolitan University, Toronto, ON, Canada
| |
Collapse
|
9
|
Rossi E, Boldrini L, Maratta MG, Gatta R, Votta C, Tortora G, Schinzari G. Radiomics to predict immunotherapy efficacy in advanced renal cell carcinoma: A retrospective study. Hum Vaccin Immunother 2023; 19:2172926. [PMID: 36723981 PMCID: PMC10012916 DOI: 10.1080/21645515.2023.2172926] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Immunotherapy has become a cornerstone for the treatment of renal cell carcinoma. Nevertheless, some patients are resistant to immune checkpoint inhibitors. The possibility to identify patients who cannot benefit from immunotherapy is a relevant clinical challenge. We analyzed the association between several radiomics features and response to immunotherapy in 53 patients treated with checkpoint inhibitors for advanced renal cell carcinoma. We found that the following features are associated with progression of disease as best tumor response: F_stat.range (p < .0004), F_stat.max (p < .0007), F_stat.var (p < .0016), F_stat.uniformity (p < .0020), F_stat.90thpercentile (p < .0050). Gross tumor volumes characterized by high values of F_stat.var and F_stat.max (greater than 60,000 and greater than 300, respectively) are most likely related to a high risk of progression. Further analyses are warranted to confirm these results. Radiomics, together with other potential predictive factors, such as gut microbiota, genetic features or circulating immune molecules, could allow a personalized treatment for patients with advanced renal cell carcinoma.
Collapse
Affiliation(s)
- Ernesto Rossi
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Luca Boldrini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Grazia Maratta
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Roberto Gatta
- Dipartimento di Scienze Cliniche e Sperimentali, Universitá degli Studi di Brescia, Brescia, Italy.,Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Claudio Votta
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giampaolo Tortora
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Schinzari
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
10
|
Shieh A, Cen SY, Varghese BA, Hwang D, Lei X, Setayesh A, Siddiqi I, Aron M, Dsouza A, Gill IS, Wallace W, Duddalwar V. Radiomics Correlation to CD68+ Tumor-Associated Macrophages in Clear Cell Renal Cell Carcinoma. Oncology 2023; 102:260-270. [PMID: 37699367 DOI: 10.1159/000534078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/04/2023] [Indexed: 09/14/2023]
Abstract
INTRODUCTION Renal cell carcinoma (RCC) is the ninth most common cancer worldwide, with clear cell RCC (ccRCC) being the most frequent histological subtype. The tumor immune microenvironment (TIME) of ccRCC is an important factor to guide treatment, but current assessments are tissue-based, which can be time-consuming and resource-intensive. In this study, we used radiomics extracted from clinically performed computed tomography (CT) as a noninvasive surrogate for CD68 tumor-associated macrophages (TAMs), a significant component of ccRCC TIME. METHODS TAM population was measured by CD68+/PanCK+ ratio and tumor-TAM clustering was measured by normalized K function calculated from multiplex immunofluorescence (mIF). A total of 1,076 regions on mIF slides from 78 patients were included. Radiomic features were extracted from multiphase CT of the ccRCC tumor. Statistical machine learning models, including random forest, Adaptive Boosting, and ElasticNet, were used to predict TAM population and tumor-TAM clustering. RESULTS The best models achieved an area under the ROC curve of 0.81 (95% CI: [0.69, 0.92]) for TAM population and 0.77 (95% CI: [0.66, 0.88]) for tumor-TAM clustering, respectively. CONCLUSION Our study demonstrates the potential of using CT radiomics-derived imaging markers as a surrogate for assessment of TAM in ccRCC for real-time treatment response monitoring and patient selection for targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Alexander Shieh
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,
| | - Steven Y Cen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Bino A Varghese
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Darryl Hwang
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Xiaomeng Lei
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ali Setayesh
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Imran Siddiqi
- Department of Pathology, University of Southern California, Los Angeles, California, USA
| | - Manju Aron
- Department of Pathology, University of Southern California, Los Angeles, California, USA
| | - Anishka Dsouza
- Division of Medical Oncology, Department of Medicine, University of Southern California, Los Angeles, California, USA
| | - Inderbir S Gill
- Institute of Urology, University of Southern California, Los Angeles, California, USA
| | - William Wallace
- Department of Pathology, University of Southern California, Los Angeles, California, USA
| | - Vinay Duddalwar
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Institute of Urology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
11
|
Saber R, Henault D, Messaoudi N, Rebolledo R, Montagnon E, Soucy G, Stagg J, Tang A, Turcotte S, Kadoury S. Radiomics using computed tomography to predict CD73 expression and prognosis of colorectal cancer liver metastases. J Transl Med 2023; 21:507. [PMID: 37501197 PMCID: PMC10375693 DOI: 10.1186/s12967-023-04175-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/30/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Finding a noninvasive radiomic surrogate of tumor immune features could help identify patients more likely to respond to novel immune checkpoint inhibitors. Particularly, CD73 is an ectonucleotidase that catalyzes the breakdown of extracellular AMP into immunosuppressive adenosine, which can be blocked by therapeutic antibodies. High CD73 expression in colorectal cancer liver metastasis (CRLM) resected with curative intent is associated with early recurrence and shorter patient survival. The aim of this study was hence to evaluate whether machine learning analysis of preoperative liver CT-scan could estimate high vs low CD73 expression in CRLM and whether such radiomic score would have a prognostic significance. METHODS We trained an Attentive Interpretable Tabular Learning (TabNet) model to predict, from preoperative CT images, stratified expression levels of CD73 (CD73High vs. CD73Low) assessed by immunofluorescence (IF) on tissue microarrays. Radiomic features were extracted from 160 segmented CRLM of 122 patients with matched IF data, preprocessed and used to train the predictive model. We applied a five-fold cross-validation and validated the performance on a hold-out test set. RESULTS TabNet provided areas under the receiver operating characteristic curve of 0.95 (95% CI 0.87 to 1.0) and 0.79 (0.65 to 0.92) on the training and hold-out test sets respectively, and outperformed other machine learning models. The TabNet-derived score, termed rad-CD73, was positively correlated with CD73 histological expression in matched CRLM (Spearman's ρ = 0.6004; P < 0.0001). The median time to recurrence (TTR) and disease-specific survival (DSS) after CRLM resection in rad-CD73High vs rad-CD73Low patients was 13.0 vs 23.6 months (P = 0.0098) and 53.4 vs 126.0 months (P = 0.0222), respectively. The prognostic value of rad-CD73 was independent of the standard clinical risk score, for both TTR (HR = 2.11, 95% CI 1.30 to 3.45, P < 0.005) and DSS (HR = 1.88, 95% CI 1.11 to 3.18, P = 0.020). CONCLUSIONS Our findings reveal promising results for non-invasive CT-scan-based prediction of CD73 expression in CRLM and warrant further validation as to whether rad-CD73 could assist oncologists as a biomarker of prognosis and response to immunotherapies targeting the adenosine pathway.
Collapse
Affiliation(s)
- Ralph Saber
- MedICAL Laboratory, Polytechnique Montréal, Montréal, H3T 1J4, Canada
- Imaging and Engineering Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis R10.430, Montréal, QC, H2X 0A9, Canada
| | - David Henault
- Cancer Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis, Room R10.430, Montréal, QC, H2X 0A9, Canada
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Service, Centre hospitalier de l'Université de Montréal, 1000, rue Saint-Denis, Montréal, QC, H2X 0C1, Canada
| | - Nouredin Messaoudi
- Cancer Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis, Room R10.430, Montréal, QC, H2X 0A9, Canada
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Service, Centre hospitalier de l'Université de Montréal, 1000, rue Saint-Denis, Montréal, QC, H2X 0C1, Canada
- Department of Surgery, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel) and Europe Hospitals, Brussels, Belgium
| | - Rolando Rebolledo
- Cancer Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis, Room R10.430, Montréal, QC, H2X 0A9, Canada
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Service, Centre hospitalier de l'Université de Montréal, 1000, rue Saint-Denis, Montréal, QC, H2X 0C1, Canada
| | - Emmanuel Montagnon
- Imaging and Engineering Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis R10.430, Montréal, QC, H2X 0A9, Canada
| | - Geneviève Soucy
- Pahology Department, Centre hospitalier de l'Université de Montréal, 1000, rue Saint-Denis, Montréal, QC, H2X 0C1, Canada
| | - John Stagg
- Cancer Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis, Room R10.430, Montréal, QC, H2X 0A9, Canada
| | - An Tang
- Imaging and Engineering Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis R10.430, Montréal, QC, H2X 0A9, Canada
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montréal, Montréal, H3T 1J4, Canada
| | - Simon Turcotte
- Cancer Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis, Room R10.430, Montréal, QC, H2X 0A9, Canada.
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Service, Centre hospitalier de l'Université de Montréal, 1000, rue Saint-Denis, Montréal, QC, H2X 0C1, Canada.
| | - Samuel Kadoury
- MedICAL Laboratory, Polytechnique Montréal, Montréal, H3T 1J4, Canada.
- Imaging and Engineering Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis R10.430, Montréal, QC, H2X 0A9, Canada.
- Department of Computer and Software Engineering, Institute of Biomedical Engineering, Polytechnique Montréal, Montréal, H3T 1J4, Canada.
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montréal, Montréal, H3T 1J4, Canada.
| |
Collapse
|
12
|
Elahmadawy MA, Ashraf A, Moustafa H, Kotb M, Abd El-Gaid S. Prognostic value of initial [ 18 F]FDG PET/computed tomography volumetric and texture analysis-based parameters in patients with head and neck squamous cell carcinoma. Nucl Med Commun 2023; 44:653-662. [PMID: 37038954 DOI: 10.1097/mnm.0000000000001695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
AIM OF WORK To determine the predictive value of initial [ 18 F]FDG PET/computed tomography (CT) volumetric and radiomics-derived analyses in patients with head and neck squamous cell carcinoma (HNSCC). METHODS Forty-six adult patients had pathologically proven HNSCC and underwent pretherapy [ 18 F]FDG PET/CT were enrolled. Semi-quantitative PET-derived volumetric [(maximum standardized uptake value (SUVmax) and mean SUV (SUVmean), total lesion glycolysis (TLG) and metabolic tumor volume (MTV)] and radiomics analyses using LIFEx 6.73.3 software were performed. RESULTS In the current study group, the receiver operating characteristic curve marked a cutoff point of 21.105 for primary MTV with area under the curve (AUC) of 0.727, sensitivity of 62.5%, and specificity of 86.8% ( P value 0.041) to distinguish responders from non-responders, while no statistically significant primary SUVmean or max or primary TLG cut off points could be determined. It also marked the cutoff point for survival prediction of 10.845 for primary MTV with AUC 0.728, sensitivity of 80%, and specificity of 77.8% ( P value 0.026). A test of the synergistic performance of PET-derived volumetric and textural features significant parameters was conducted in an attempt to develop the most accurate and stable prediction model. Therefore, multivariate logistic regression analysis was performed to detect independent predictors of mortality. With a high specificity of 97.1% and an overall accuracy of 89.1%, the combination of primary tumor MTV and the textural feature gray-level co-occurrence matrix correlation provided the most accurate prediction of mortality ( P value < 0.001). CONCLUSION Textural feature indices are a noninvasive method for capturing intra-tumoral heterogeneity. In our study, a PET-derived prediction model was successfully generated with high specificity and accuracy.
Collapse
Affiliation(s)
| | - Aya Ashraf
- Nuclear Medicine Unit, National Cancer Institute
| | - Hosna Moustafa
- Nuclear Medicine Unit, Kasr Al-Ainy (NEMROCK Center), Cairo University, Cairo, Egypt
| | - Magdy Kotb
- Nuclear Medicine Unit, National Cancer Institute
| | | |
Collapse
|
13
|
Khalili N, Kazerooni AF, Familiar A, Haldar D, Kraya A, Foster J, Koptyra M, Storm PB, Resnick AC, Nabavizadeh A. Radiomics for characterization of the glioma immune microenvironment. NPJ Precis Oncol 2023; 7:59. [PMID: 37337080 DOI: 10.1038/s41698-023-00413-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/02/2023] [Indexed: 06/21/2023] Open
Abstract
Increasing evidence suggests that besides mutational and molecular alterations, the immune component of the tumor microenvironment also substantially impacts tumor behavior and complicates treatment response, particularly to immunotherapies. Although the standard method for characterizing tumor immune profile is through performing integrated genomic analysis on tissue biopsies, the dynamic change in the immune composition of the tumor microenvironment makes this approach not feasible, especially for brain tumors. Radiomics is a rapidly growing field that uses advanced imaging techniques and computational algorithms to extract numerous quantitative features from medical images. Recent advances in machine learning methods are facilitating biological validation of radiomic signatures and allowing them to "mine" for a variety of significant correlates, including genetic, immunologic, and histologic data. Radiomics has the potential to be used as a non-invasive approach to predict the presence and density of immune cells within the microenvironment, as well as to assess the expression of immune-related genes and pathways. This information can be essential for patient stratification, informing treatment decisions and predicting patients' response to immunotherapies. This is particularly important for tumors with difficult surgical access such as gliomas. In this review, we provide an overview of the glioma microenvironment, describe novel approaches for clustering patients based on their tumor immune profile, and discuss the latest progress on utilization of radiomics for immune profiling of glioma based on current literature.
Collapse
Affiliation(s)
- Nastaran Khalili
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anahita Fathi Kazerooni
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ariana Familiar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Debanjan Haldar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Kraya
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jessica Foster
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mateusz Koptyra
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Phillip B Storm
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adam C Resnick
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ali Nabavizadeh
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Ng J, Gregucci F, Pennell RT, Nagar H, Golden EB, Knisely JPS, Sanfilippo NJ, Formenti SC. MRI-LINAC: A transformative technology in radiation oncology. Front Oncol 2023; 13:1117874. [PMID: 36776309 PMCID: PMC9911688 DOI: 10.3389/fonc.2023.1117874] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/16/2023] [Indexed: 01/28/2023] Open
Abstract
Advances in radiotherapy technologies have enabled more precise target guidance, improved treatment verification, and greater control and versatility in radiation delivery. Amongst the recent novel technologies, Magnetic Resonance Imaging (MRI) guided radiotherapy (MRgRT) may hold the greatest potential to improve the therapeutic gains of image-guided delivery of radiation dose. The ability of the MRI linear accelerator (LINAC) to image tumors and organs with on-table MRI, to manage organ motion and dose delivery in real-time, and to adapt the radiotherapy plan on the day of treatment while the patient is on the table are major advances relative to current conventional radiation treatments. These advanced techniques demand efficient coordination and communication between members of the treatment team. MRgRT could fundamentally transform the radiotherapy delivery process within radiation oncology centers through the reorganization of the patient and treatment team workflow process. However, the MRgRT technology currently is limited by accessibility due to the cost of capital investment and the time and personnel allocation needed for each fractional treatment and the unclear clinical benefit compared to conventional radiotherapy platforms. As the technology evolves and becomes more widely available, we present the case that MRgRT has the potential to become a widely utilized treatment platform and transform the radiation oncology treatment process just as earlier disruptive radiation therapy technologies have done.
Collapse
Affiliation(s)
- John Ng
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States,*Correspondence: John Ng,
| | - Fabiana Gregucci
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States,Department of Radiation Oncology, Miulli General Regional Hospital, Acquaviva delle Fonti, Bari, Italy
| | - Ryan T. Pennell
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Himanshu Nagar
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Encouse B. Golden
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| | | | | | - Silvia C. Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
15
|
Li S, Zhou B. A review of radiomics and genomics applications in cancers: the way towards precision medicine. Radiat Oncol 2022; 17:217. [PMID: 36585716 PMCID: PMC9801589 DOI: 10.1186/s13014-022-02192-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
The application of radiogenomics in oncology has great prospects in precision medicine. Radiogenomics combines large volumes of radiomic features from medical digital images, genetic data from high-throughput sequencing, and clinical-epidemiological data into mathematical modelling. The amalgamation of radiomics and genomics provides an approach to better study the molecular mechanism of tumour pathogenesis, as well as new evidence-supporting strategies to identify the characteristics of cancer patients, make clinical decisions by predicting prognosis, and improve the development of individualized treatment guidance. In this review, we summarized recent research on radiogenomics applications in solid cancers and presented the challenges impeding the adoption of radiomics in clinical practice. More standard guidelines are required to normalize radiomics into reproducible and convincible analyses and develop it as a mature field.
Collapse
Affiliation(s)
- Simin Li
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Hospital of China Medical University, Shenyang, 110001 Liaoning People’s Republic of China
| | - Baosen Zhou
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Hospital of China Medical University, Shenyang, 110001 Liaoning People’s Republic of China
| |
Collapse
|
16
|
Mohammadi A, Mirza-Aghazadeh-Attari M, Faeghi F, Homayoun H, Abolghasemi J, Vogl TJ, Bureau NJ, Bakhshandeh M, Acharya RU, Abbasian Ardakani A. Tumor Microenvironment, Radiology, and Artificial Intelligence: Should We Consider Tumor Periphery? JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2022; 41:3079-3090. [PMID: 36000351 DOI: 10.1002/jum.16086] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVES The tumor microenvironment (TME) consists of cellular and noncellular components which enable the tumor to interact with its surroundings and plays an important role in the tumor progression and how the immune system reacts to the malignancy. In the present study, we investigate the diagnostic potential of the TME in differentiating benign and malignant lesions using image quantification and machine learning. METHODS A total of 229 breast lesions and 220 cervical lymph nodes were included in the study. A group of expert radiologists first performed medical imaging and segmented the lesions, after which a rectangular mask was drawn, encompassing all of the contouring. The mask was extended in each axis up to 50%, and 29 radiomics features were extracted from each mask. Radiomics features that showed a significant difference in each contour were used to develop a support vector machine (SVM) classifier for benign and malignant lesions in breast and lymph node images separately. RESULTS Single radiomics features extracted from extended contours outperformed radiologists' contours in both breast and lymph node lesions. Furthermore, when fed into the SVM model, the extended models also outperformed the radiologist's contour, achieving an area under the receiver operating characteristic curve of 0.887 and 0.970 in differentiating breast and lymph node lesions, respectively. CONCLUSIONS Our results provide convincing evidence regarding the importance of the tumor periphery and TME in medical imaging diagnosis. We propose that the immediate tumor periphery should be considered for differentiating benign and malignant lesions in image quantification studies.
Collapse
Affiliation(s)
- Afshin Mohammadi
- Department of Radiology, Faculty of Medicine, Urmia University of Medical Science, Urmia, Iran
| | | | - Fariborz Faeghi
- Department of Radiology Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hasan Homayoun
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamileh Abolghasemi
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Thomas J Vogl
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Nathalie J Bureau
- Department of Radiology, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | - Mohsen Bakhshandeh
- Department of Radiology Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rajendra U Acharya
- Department of Electronics and Computer Engineering, Ngee Ann Polytechnic, Singapore
- Department of Biomedical Engineering, School of Science and Technology, SUSS University, Singapore
- Department of Biomedical Informatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Ali Abbasian Ardakani
- Department of Radiology Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Crombé A, Roulleau‐Dugage M, Italiano A. The diagnosis, classification, and treatment of sarcoma in this era of artificial intelligence and immunotherapy. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 42:1288-1313. [PMID: 36260064 PMCID: PMC9759765 DOI: 10.1002/cac2.12373] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 09/20/2022] [Accepted: 10/08/2022] [Indexed: 01/25/2023]
Abstract
Soft-tissue sarcomas (STS) represent a group of rare and heterogeneous tumors associated with several challenges, including incorrect or late diagnosis, the lack of clinical expertise, and limited therapeutic options. Digital pathology and radiomics represent transformative technologies that appear promising for improving the accuracy of cancer diagnosis, characterization and monitoring. Herein, we review the potential role of the application of digital pathology and radiomics in managing patients with STS. We have particularly described the main results and the limits of the studies using radiomics to refine diagnosis or predict the outcome of patients with soft-tissue sarcomas. We also discussed the current limitation of implementing radiomics in routine settings. Standard management approaches for STS have not improved since the early 1970s. Immunotherapy has revolutionized cancer treatment; nonetheless, immuno-oncology agents have not yet been approved for patients with STS. However, several lines of evidence indicate that immunotherapy may represent an efficient therapeutic strategy for this group of diseases. Thus, we emphasized the remarkable potential of immunotherapy in sarcoma treatment by focusing on recent data regarding the immune landscape of these tumors. We have particularly emphasized the fact that the development of immunotherapy for sarcomas is not an aspect of histology (except for alveolar soft-part sarcoma) but rather that of the tumor microenvironment. Future studies investigating immunotherapy strategies in sarcomas should incorporate at least the presence of tertiary lymphoid structures as a stratification factor in their design, besides including a strong translational program that will allow for a better understanding of the determinants involved in sensitivity and treatment resistance to immune-oncology agents.
Collapse
Affiliation(s)
- Amandine Crombé
- Department of ImagingInstitut BergoniéBordeauxNouvelle‐AquitaineF‐33076France,Faculty of MedicineUniversity of BordeauxBordeauxNouvelle‐AquitaineF‐33000France
| | | | - Antoine Italiano
- Faculty of MedicineUniversity of BordeauxBordeauxNouvelle‐AquitaineF‐33000France,Early Phase Trials and Sarcoma UnitInstitut BergoniéBordeauxNouvelle‐AquitaineF‐33076France
| |
Collapse
|
18
|
Kobayashi T. RadiomicsJ: a library to compute radiomic features. Radiol Phys Technol 2022; 15:255-263. [PMID: 35792994 DOI: 10.1007/s12194-022-00664-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 10/17/2022]
Abstract
Despite the widely recognized need for radiomics research, the development and use of full-scale radiomics-based predictive models in clinical practice remains scarce. This is because of the lack of well-established methodologies for radiomic research and the need to develop systems to support radiomic feature calculations and predictive model use. Several excellent programs for calculating radiomic features have been developed. However, there are still issues such as the types of image features, variations in the calculated results, and the limited system environment in which to run the program. Against this background, we developed RadiomicsJ, an open-source radiomic feature computation library. RadiomicsJ will not only be a new research tool to enhance the efficiency of radiomics research but will also become a knowledge resource for medical imaging feature studies through its release as an open-source program.
Collapse
Affiliation(s)
- Tatsuaki Kobayashi
- Visionary Imaging Services, Inc, 1-16-19, Nagatsuta, Midori-Ku, Yokohama, Kanagawa, Japan.
| |
Collapse
|
19
|
Bekker RA, Zahid MU, Binning JM, Spring BQ, Hwu P, Pilon-Thomas S, Enderling H. Rethinking the immunotherapy numbers game. J Immunother Cancer 2022; 10:jitc-2022-005107. [PMID: 35793871 PMCID: PMC9260835 DOI: 10.1136/jitc-2022-005107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2022] [Indexed: 11/23/2022] Open
Abstract
Immunotherapies are a major breakthrough in oncology, yielding unprecedented response rates for some cancers. Especially in combination with conventional treatments or targeted agents, immunotherapeutics offer invaluable tools to improve outcomes for many patients. However, why not all patients have a favorable response remains unclear. There is an increasing appreciation of the contributions of the complex tumor microenvironment, and the tumor-immune ecosystem in particular, to treatment outcome. To date, however, there exists no immune biomarker to explain why two patients with similar clinical stage and molecular profile would have different treatment outcomes. We hypothesize that it is critical to understand both the immune and tumor states to understand how the complex system will respond to treatment. Here, we present how integrated mathematical oncology approaches can help conceptualize the effect of various immunotherapies on a patient’s tumor and local immune environment, and how combinations of immunotherapy and cytotoxic therapy may be used to improve tumor response and control and limit toxicity on a per patient basis.
Collapse
Affiliation(s)
- Rebecca A Bekker
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA.,Cancer Biology Ph.D. Program, University of South Florida, Tampa, Florida, USA
| | - Mohammad U Zahid
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Jennifer M Binning
- Department of Molecular Oncology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Bryan Q Spring
- Translational Biophotonics Cluster, Northeastern University, Boston, Massachusetts, USA.,Department of Physics, Northeastern University, Boston, Massachusetts, USA.,Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | | | - Shari Pilon-Thomas
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Heiko Enderling
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA .,Department of Radiation Oncology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| |
Collapse
|
20
|
Manafi-Farid R, Askari E, Shiri I, Pirich C, Asadi M, Khateri M, Zaidi H, Beheshti M. [ 18F]FDG-PET/CT radiomics and artificial intelligence in lung cancer: Technical aspects and potential clinical applications. Semin Nucl Med 2022; 52:759-780. [PMID: 35717201 DOI: 10.1053/j.semnuclmed.2022.04.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/10/2022] [Accepted: 04/13/2022] [Indexed: 02/07/2023]
Abstract
Lung cancer is the second most common cancer and the leading cause of cancer-related death worldwide. Molecular imaging using [18F]fluorodeoxyglucose Positron Emission Tomography and/or Computed Tomography ([18F]FDG-PET/CT) plays an essential role in the diagnosis, evaluation of response to treatment, and prediction of outcomes. The images are evaluated using qualitative and conventional quantitative indices. However, there is far more information embedded in the images, which can be extracted by sophisticated algorithms. Recently, the concept of uncovering and analyzing the invisible data extracted from medical images, called radiomics, is gaining more attention. Currently, [18F]FDG-PET/CT radiomics is growingly evaluated in lung cancer to discover if it enhances the diagnostic performance or implication of [18F]FDG-PET/CT in the management of lung cancer. In this review, we provide a short overview of the technical aspects, as they are discussed in different articles of this special issue. We mainly focus on the diagnostic performance of the [18F]FDG-PET/CT-based radiomics and the role of artificial intelligence in non-small cell lung cancer, impacting the early detection, staging, prediction of tumor subtypes, biomarkers, and patient's outcomes.
Collapse
Affiliation(s)
- Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Emran Askari
- Department of Nuclear Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - Christian Pirich
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Mahboobeh Asadi
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maziar Khateri
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland; Geneva University Neurocenter, Geneva University, Geneva, Switzerland; Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
21
|
Rinaldi L, Pezzotta F, Santaniello T, De Marco P, Bianchini L, Origgi D, Cremonesi M, Milani P, Mariani M, Botta F. HeLLePhant: A phantom mimicking non-small cell lung cancer for texture analysis in CT images. Phys Med 2022; 97:13-24. [PMID: 35334407 DOI: 10.1016/j.ejmp.2022.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/01/2022] [Accepted: 03/14/2022] [Indexed: 01/06/2023] Open
Abstract
PURPOSE Phantoms mimicking human tissue heterogeneity and intensity are required to establish radiomic features robustness in Computed Tomography (CT) images. We developed inserts with two different techniques for the radiomic study of Non-Small Cell Lung Cancer (NSCLC) lesions. METHODS We developed two insert prototypes: two 3D-printed made of glycol-modified polyethylene terephthalate (PET-G), and nine with sodium polyacrylate plus iodinated contrast medium. The inserts were put in a handcraft phantom (HeLLePhant). We also analysed four materials of a commercial homogeneous phantom (Catphan® 424) and collected 29 NSCLC patients for comparison. All the CT acquisitions were performed with the same clinical protocol and scanner at 120kVp. The HeLLePhant phantom was scanned ten times in fixed condition at 120kVp and 100kVp for repeatability investigation. We extracted 153 radiomic features using Pyradiomics. To compare the features between phantoms and patients, we computed how many phantom features fell in the range between 10th and 90th percentile of the corresponding patient values. We deemed repeatable the features with a coefficient of variation (CV) less than or equal to 0.10. RESULTS The best similarity with the patients was obtained with the polyacrylate inserts (55.6-90.2%), the worst with Catphan (15.7-19.0%). For the PET-G inserts 35.3% and 36.6% of the features match the patient range. We found high repeatability for all the inserts of the HeLLePhant phantom (74.3-100% at 120kVp, 75.7-97.9% at 100kVp), and observed a texture dependency in repeatability. CONCLUSIONS Our study shows a promising way to construct heterogeneous inserts mimicking a target tissue for radiomic studies.
Collapse
Affiliation(s)
- Lisa Rinaldi
- Department of Physics, Università degli Studi di Pavia and INFN, via Bassi 6, 27100 Pavia, Italy; Radiation Research Unit, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy.
| | - Federico Pezzotta
- CIMaINa, Department of Physics, Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Tommaso Santaniello
- CIMaINa, Department of Physics, Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Paolo De Marco
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Linda Bianchini
- Department of Physics, Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Daniela Origgi
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Marta Cremonesi
- Radiation Research Unit, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Paolo Milani
- CIMaINa, Department of Physics, Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Manuel Mariani
- Department of Physics, Università degli Studi di Pavia and INFN, via Bassi 6, 27100 Pavia, Italy
| | - Francesca Botta
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| |
Collapse
|
22
|
The Role of Radiomics in the Era of Immune Checkpoint Inhibitors: A New Protagonist in the Jungle of Response Criteria. J Clin Med 2022; 11:jcm11061740. [PMID: 35330068 PMCID: PMC8948743 DOI: 10.3390/jcm11061740] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/08/2022] [Accepted: 03/18/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The introduction of immune checkpoint inhibitors has represented a milestone in cancer treatment. Despite PD-L1 expression being the standard biomarker used before the start of therapy, there is still a strict need to identify complementary non-invasive biomarkers in order to better select patients. In this context, radiomics is an emerging approach for examining medical images and clinical data by capturing multiple features hidden from human eye and is potentially able to predict response assessment and survival in the course of immunotherapy. We reviewed the available studies investigating the role of radiomics in cancer patients, focusing on non-small cell lung cancer treated with immune checkpoint inhibitors. Although preliminary research shows encouraging results, different issues need to be solved before radiomics can enter into clinical practice. Abstract Immune checkpoint inhibitors (ICI) have demonstrated encouraging results in terms of durable clinical benefit and survival in several malignancies. Nevertheless, the search to identify an “ideal” biomarker for predicting response to ICI is still far from over. Radiomics is a new translational field of study aiming to extract, by dedicated software, several features from a given medical image, ranging from intensity distribution and spatial heterogeneity to higher-order statistical parameters. Based on these premises, our review aims to summarize the current status of radiomics as a potential predictor of clinical response following immunotherapy treatment. A comprehensive search of PubMed results was conducted. All studies published in English up to and including December 2021 were selected, comprising those that explored computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET) for radiomic analyses in the setting of ICI. Several studies have demonstrated the potential applicability of radiomic features in the monitoring of the therapeutic response beyond the traditional morphologic and metabolic criteria, as well as in the prediction of survival or non-invasive assessment of the tumor microenvironment. Nevertheless, important limitations emerge from our review in terms of standardization in feature selection, data sharing, and methods, as well as in external validation. Additionally, there is still need for prospective clinical trials to confirm the potential significant role of radiomics during immunotherapy.
Collapse
|
23
|
Immunotherapeutic Approaches for Glioblastoma Treatment. Biomedicines 2022; 10:biomedicines10020427. [PMID: 35203636 PMCID: PMC8962267 DOI: 10.3390/biomedicines10020427] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma remains a challenging disease to treat, despite well-established standard-of-care treatments, with a median survival consistently of less than 2 years. In this review, we delineate the unique disease-specific challenges for immunotherapies, both brain-related and non-brain-related, which will need to be adequately overcome for the development of effective treatments. We also review current immunotherapy treatments, with a focus on clinical applications, and propose future directions for the field of GBM immunotherapy.
Collapse
|
24
|
Farwell MD, Mankoff DA. Analysis of Routine Computed Tomographic Scans With Radiomics and Machine Learning: One Step Closer to Clinical Practice. JAMA Oncol 2022; 8:393-394. [PMID: 35050318 DOI: 10.1001/jamaoncol.2021.6768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Michael D Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - David A Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
25
|
Kang CY, Duarte SE, Kim HS, Kim E, Park J, Lee AD, Kim Y, Kim L, Cho S, Oh Y, Gim G, Park I, Lee D, Abazeed M, Velichko YS, Chae YK. OUP accepted manuscript. Oncologist 2022; 27:e471-e483. [PMID: 35348765 PMCID: PMC9177100 DOI: 10.1093/oncolo/oyac036] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 01/14/2022] [Indexed: 11/17/2022] Open
Abstract
The recent, rapid advances in immuno-oncology have revolutionized cancer treatment and spurred further research into tumor biology. Yet, cancer patients respond variably to immunotherapy despite mounting evidence to support its efficacy. Current methods for predicting immunotherapy response are unreliable, as these tests cannot fully account for tumor heterogeneity and microenvironment. An improved method for predicting response to immunotherapy is needed. Recent studies have proposed radiomics—the process of converting medical images into quantitative data (features) that can be processed using machine learning algorithms to identify complex patterns and trends—for predicting response to immunotherapy. Because patients undergo numerous imaging procedures throughout the course of the disease, there exists a wealth of radiological imaging data available for training radiomics models. And because radiomic features reflect cancer biology, such as tumor heterogeneity and microenvironment, these models have enormous potential to predict immunotherapy response more accurately than current methods. Models trained on preexisting biomarkers and/or clinical outcomes have demonstrated potential to improve patient stratification and treatment outcomes. In this review, we discuss current applications of radiomics in oncology, followed by a discussion on recent studies that use radiomics to predict immunotherapy response and toxicity.
Collapse
Affiliation(s)
| | | | - Hye Sung Kim
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eugene Kim
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Alice Daeun Lee
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yeseul Kim
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Leeseul Kim
- Department of Internal Medicine, AMITA Health Saint Francis Hospital, Evanston, IL, USA
| | - Sukjoo Cho
- Department of Pediatrics, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Yoojin Oh
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Gahyun Gim
- Department of Hematology and Oncology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Inae Park
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Dongyup Lee
- Department of Physical Medicine and Rehabilitation, Geisinger Health System, Danville, PA, USA
| | - Mohamed Abazeed
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yury S Velichko
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Young Kwang Chae
- Corresponding author: Young Kwang Chae, Department of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
26
|
Guglielmo P, Marturano F, Bettinelli A, Gregianin M, Paiusco M, Evangelista L. Additional Value of PET Radiomic Features for the Initial Staging of Prostate Cancer: A Systematic Review from the Literature. Cancers (Basel) 2021; 13:cancers13236026. [PMID: 34885135 PMCID: PMC8657371 DOI: 10.3390/cancers13236026] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Prostate cancer (PCa) is one of the most frequent malignancies diagnosed in men and its prognosis depends on the stage at diagnosis. Molecular imaging, namely PET/CT or PET/MRI using prostate-specific radiotracers, has gained increasing application in accurately evaluating PCa at staging, especially in cases of high-risk disease, and it is now also recommended by international guidelines. Radiomic analysis is an emerging research field with a high potential to offer non-invasive and longitudinal biomarkers for personalized medicine, and several applications have been described in oncology patients. In this review, we discuss the available evidence on the role of radiomic analysis in PCa imaging at staging, exploring two different hybrid imaging modalities, such as PET/CT and PET/MRI, and the whole spectrum of radiotracers involved. Abstract We performed a systematic review of the literature to provide an overview of the application of PET radiomics for the prediction of the initial staging of prostate cancer (PCa), and to discuss the additional value of radiomic features over clinical data. The most relevant databases and web sources were interrogated by using the query “prostate AND radiomic* AND PET”. English-language original articles published before July 2021 were considered. A total of 28 studies were screened for eligibility and 6 of them met the inclusion criteria and were, therefore, included for further analysis. All studies were based on human patients. The average number of patients included in the studies was 72 (range 52–101), and the average number of high-order features calculated per study was 167 (range 50–480). The radiotracers used were [68Ga]Ga-PSMA-11 (in four out of six studies), [18F]DCFPyL (one out of six studies), and [11C]Choline (one out of six studies). Considering the imaging modality, three out of six studies used a PET/CT scanner and the other half a PET/MRI tomograph. Heterogeneous results were reported regarding radiomic methods (e.g., segmentation modality) and considered features. The studies reported several predictive markers including first-, second-, and high-order features, such as “kurtosis”, “grey-level uniformity”, and “HLL wavelet mean”, respectively, as well as PET-based metabolic parameters. The strengths and weaknesses of PET radiomics in this setting of disease will be largely discussed and a critical analysis of the available data will be reported. In our review, radiomic analysis proved to add useful information for lesion detection and the prediction of tumor grading of prostatic lesions, even when they were missed at visual qualitative assessment due to their small size; furthermore, PET radiomics could play a synergistic role with the mpMRI radiomic features in lesion evaluation. The most common limitations of the studies were the small sample size, retrospective design, lack of validation on external datasets, and unavailability of univocal cut-off values for the selected radiomic features.
Collapse
Affiliation(s)
- Priscilla Guglielmo
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV—IRCCS, 31033 Castelfranco Veneto, Italy; (P.G.); (M.G.)
| | - Francesca Marturano
- Medical Physics Unit, Veneto Institute of Oncology IOV—IRCCS, 32168 Padova, Italy; (F.M.); (A.B.); (M.P.)
| | - Andrea Bettinelli
- Medical Physics Unit, Veneto Institute of Oncology IOV—IRCCS, 32168 Padova, Italy; (F.M.); (A.B.); (M.P.)
| | - Michele Gregianin
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV—IRCCS, 31033 Castelfranco Veneto, Italy; (P.G.); (M.G.)
| | - Marta Paiusco
- Medical Physics Unit, Veneto Institute of Oncology IOV—IRCCS, 32168 Padova, Italy; (F.M.); (A.B.); (M.P.)
| | - Laura Evangelista
- Nuclear Medicine Unit, Department of Medicine DIMED, University of Padova, 32168 Padova, Italy
- Correspondence: ; Tel.: +39-0498211310; Fax: +39-0498213008
| |
Collapse
|
27
|
Rebuzzi SE, Banna GL, Murianni V, Damassi A, Giunta EF, Fraggetta F, De Giorgi U, Cathomas R, Rescigno P, Brunelli M, Fornarini G. Prognostic and Predictive Factors in Advanced Urothelial Carcinoma Treated with Immune Checkpoint Inhibitors: A Review of the Current Evidence. Cancers (Basel) 2021; 13:5517. [PMID: 34771680 PMCID: PMC8583566 DOI: 10.3390/cancers13215517] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022] Open
Abstract
In recent years, the treatment landscape of urothelial carcinoma has significantly changed due to the introduction of immune checkpoint inhibitors (ICIs), which are the standard of care for second-line treatment and first-line platinum-ineligible patients with advanced disease. Despite the overall survival improvement, only a minority of patients benefit from this immunotherapy. Therefore, there is an unmet need to identify prognostic and predictive biomarkers or models to select patients who will benefit from ICIs, especially in view of novel therapeutic agents. This review describes the prognostic and predictive role, and clinical readiness, of clinical and tumour factors, including new molecular classes, tumour mutational burden, mutational signatures, circulating tumour DNA, programmed death-ligand 1, inflammatory indices and clinical characteristics for patients with urothelial cancer treated with ICIs. A classification of these factors according to the levels of evidence and grades of recommendation currently indicates both a prognostic and predictive value for ctDNA and a prognostic relevance only for concomitant medications and patients' characteristics.
Collapse
Affiliation(s)
- Sara Elena Rebuzzi
- Medical Oncology, Ospedale San Paolo, 17100 Savona, Italy
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, 16132 Genova, Italy
| | | | - Veronica Murianni
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (V.M.); (G.F.)
| | - Alessandra Damassi
- Academic Unit of Medical Oncology, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Emilio Francesco Giunta
- Department of Precision Medicine, Università Degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy;
| | | | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Richard Cathomas
- Division of Oncology/Hematology, Kantonsspital Graubünden, 7000 Chur, Switzerland;
| | - Pasquale Rescigno
- Interdisciplinary Group for Translational Research and Clinical Trials, Urogenital Cancers GIRT-Uro, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy;
| | - Matteo Brunelli
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Giuseppe Fornarini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (V.M.); (G.F.)
| |
Collapse
|