1
|
Ahmed TM, Zhu Z, Yasrab M, Blanco A, Kawamoto S, He J, Fishman EK, Chu L, Javed AA. Preoperative Prediction of Lymph Node Metastases in Nonfunctional Pancreatic Neuroendocrine Tumors Using a Combined CT Radiomics-Clinical Model. Ann Surg Oncol 2024; 31:8136-8145. [PMID: 39179862 DOI: 10.1245/s10434-024-16064-4] [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: 05/10/2024] [Accepted: 08/04/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND PanNETs are a rare group of pancreatic tumors that display heterogeneous histopathological and clinical behavior. Nodal disease has been established as one of the strongest predictors of patient outcomes in PanNETs. Lack of accurate preoperative assessment of nodal disease is a major limitation in the management of these patients, in particular those with small (< 2 cm) low-grade tumors. The aim of the study was to evaluate the ability of radiomic features (RF) to preoperatively predict the presence of nodal disease in pancreatic neuroendocrine tumors (PanNETs). PATIENTS AND METHODS An institutional database was used to identify patients with nonfunctional PanNETs undergoing resection. Pancreas protocol computed tomography was obtained, manually segmented, and RF were extracted. These were analyzed using the minimum redundancy maximum relevance analysis for hierarchical feature selection. Youden index was used to identify the optimal cutoff for predicting nodal disease. A random forest prediction model was trained using RF and clinicopathological characteristics and validated internally. RESULTS Of the 320 patients included in the study, 92 (28.8%) had nodal disease based on histopathological assessment of the surgical specimen. A radiomic signature based on ten selected RF was developed. Clinicopathological characteristics predictive of nodal disease included tumor grade and size. Upon internal validation the combined radiomics and clinical feature model demonstrated adequate performance (AUC 0.80) in identifying nodal disease. The model accurately identified nodal disease in 85% of patients with small tumors (< 2 cm). CONCLUSIONS Non-invasive preoperative assessment of nodal disease using RF and clinicopathological characteristics is feasible.
Collapse
Affiliation(s)
- Taha M Ahmed
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Zhuotun Zhu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Mohammad Yasrab
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Alejandra Blanco
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Jin He
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Linda Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Ammar A Javed
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, NYU Langone Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
2
|
van den Ende T, Kuijper SC, Widaatalla Y, Noortman WA, van Velden FHP, Woodruff HC, van der Pol Y, Moldovan N, Pegtel DM, Derks S, Bijlsma MF, Mouliere F, de Geus-Oei PLF, Lambin PP, van Laarhoven PHWM. Integrating Clinical Variables, Radiomics, and Tumor-derived Cell-free DNA for Enhanced Prediction of Resectable Esophageal Adenocarcinoma Outcomes. Int J Radiat Oncol Biol Phys 2024:S0360-3016(24)03468-0. [PMID: 39424077 DOI: 10.1016/j.ijrobp.2024.10.010] [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: 03/25/2024] [Revised: 09/13/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND The value of integrating clinical variables, radiomics, and tumor-derived cell-free DNA (cfDNA) for the prediction of survival and response to chemoradiation of resectable esophageal adenocarcinoma (rEAC) patients is not yet known. Our aim was to investigate if radiomics and cfDNA metrics combined with clinical variables can improve personalized predictions. METHODS A cohort of 111 rEAC patients from two centers treated with neoadjuvant chemoradiotherapy was used for exploratory retrospective analyses. Models combining the clinical variables of the SOURCE survival model with radiomic features and cfDNA, were built using elastic net regression and internally validated using 5-fold cross validation. Model performance for overall survival (OS) and time to progression (TTP) were evaluated with the C-index and the area under the curve (AUC) for pathological complete response (pCR) RESULTS: The best performing baseline models for OS and TTP were based on the combination of SOURCE-cfDNA which reached a C-index of 0.55 and 0.59 compared to 0.44-0.45 with SOURCE alone. The addition of re-staging PET radiomics to SOURCE was the most promising addition for predicting OS (C-index: 0.65) and TTP (C-index: 0.60). Baseline risk-stratification was achieved for OS and TTP by combining SOURCE with radiomics or cfDNA, log-rank p<0.01. The best performing combination model for the prediction of pCR reached an AUC of 0.61 compared to 0.47 with SOURCE variables alone. CONCLUSIONS The addition of radiomics and cfDNA can improve the performance of an established survival model. External validity needs to be further assessed in future studies together with the optimization of radiomic pipelines.
Collapse
Affiliation(s)
- Tom van den Ende
- Amsterdam UMC, University of Amsterdam, Department of Medical Oncology, Meibergdreef 9, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Steven C Kuijper
- Amsterdam UMC, University of Amsterdam, Department of Medical Oncology, Meibergdreef 9, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| | - Yousif Widaatalla
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Wyanne A Noortman
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands; TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Floris H P van Velden
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Henry C Woodruff
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6200 Maastricht, The Netherlands
| | - Ymke van der Pol
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Norbert Moldovan
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, De Boelelaan 1117, Amsterdam, the Netherlands
| | - D Michiel Pegtel
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Sarah Derks
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Oncology, De Boelelaan 1117, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Maarten F Bijlsma
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Oncode Institute, Utrecht, the Netherlands; Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, Amsterdam, the Netherlands
| | - Florent Mouliere
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Prof Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands; TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands; Department of Radiation Science & Technology, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Prof Philippe Lambin
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6200 Maastricht, The Netherlands
| | - Prof Hanneke W M van Laarhoven
- Amsterdam UMC, University of Amsterdam, Department of Medical Oncology, Meibergdreef 9, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Kocak B, Akinci D'Antonoli T, Ates Kus E, Keles A, Kala A, Kose F, Kadioglu M, Solak S, Sunman S, Temiz ZH. Self-reported checklists and quality scoring tools in radiomics: a meta-research. Eur Radiol 2024; 34:5028-5040. [PMID: 38180530 DOI: 10.1007/s00330-023-10487-5] [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: 09/24/2023] [Revised: 11/11/2023] [Accepted: 11/24/2023] [Indexed: 01/06/2024]
Abstract
OBJECTIVE To evaluate the use of reporting checklists and quality scoring tools for self-reporting purposes in radiomics literature. METHODS Literature search was conducted in PubMed (date, April 23, 2023). The radiomics literature was sampled at random after a sample size calculation with a priori power analysis. A systematic assessment for self-reporting, including the use of documentation such as completed checklists or quality scoring tools, was conducted in original research papers. These eligible papers underwent independent evaluation by a panel of nine readers, with three readers assigned to each paper. Automatic annotation was used to assist in this process. Then, a detailed item-by-item confirmation analysis was carried out on papers with checklist documentation, with independent evaluation of two readers. RESULTS The sample size calculation yielded 117 papers. Most of the included papers were retrospective (94%; 110/117), single-center (68%; 80/117), based on their private data (89%; 104/117), and lacked external validation (79%; 93/117). Only seven papers (6%) had at least one self-reported document (Radiomics Quality Score (RQS), Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis (TRIPOD), or Checklist for Artificial Intelligence in Medical Imaging (CLAIM)), with a statistically significant binomial test (p < 0.001). Median rate of confirmed items for all three documents was 81% (interquartile range, 6). For quality scoring tools, documented scores were higher than suggested scores, with a mean difference of - 7.2 (standard deviation, 6.8). CONCLUSION Radiomic publications often lack self-reported checklists or quality scoring tools. Even when such documents are provided, it is essential to be cautious, as the accuracy of the reported items or scores may be questionable. CLINICAL RELEVANCE STATEMENT Current state of radiomic literature reveals a notable absence of self-reporting with documentation and inaccurate reporting practices. This critical observation may serve as a catalyst for motivating the radiomics community to adopt and utilize such tools appropriately, thereby fostering rigor, transparency, and reproducibility of their research, moving the field forward. KEY POINTS • In radiomics literature, there has been a notable absence of self-reporting with documentation. • Even if such documents are provided, it is critical to exercise caution because the accuracy of the reported items or scores may be questionable. • Radiomics community needs to be motivated to adopt and appropriately utilize the reporting checklists and quality scoring tools.
Collapse
Affiliation(s)
- Burak Kocak
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, 34480, Turkey.
| | - Tugba Akinci D'Antonoli
- Institute of Radiology and Nuclear Medicine, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Ece Ates Kus
- Department of Neuroradiology, Klinikum Lippe, Lemgo, Germany
| | - Ali Keles
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, 34480, Turkey
| | - Ahmet Kala
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, 34480, Turkey
| | - Fadime Kose
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, 34480, Turkey
| | - Mehmet Kadioglu
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, 34480, Turkey
| | - Sila Solak
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, 34480, Turkey
| | - Seyma Sunman
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, 34480, Turkey
| | - Zisan Hayriye Temiz
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, 34480, Turkey
| |
Collapse
|
4
|
Collarino A, Feudo V, Pasciuto T, Florit A, Pfaehler E, de Summa M, Bizzarri N, Annunziata S, Zannoni GF, de Geus-Oei LF, Ferrandina G, Gambacorta MA, Scambia G, Boellaard R, Sala E, Rufini V, van Velden FH. Is PET Radiomics Useful to Predict Pathologic Tumor Response and Prognosis in Locally Advanced Cervical Cancer? J Nucl Med 2024; 65:962-970. [PMID: 38548352 DOI: 10.2967/jnumed.123.267044] [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: 11/10/2023] [Revised: 03/15/2024] [Indexed: 06/05/2024] Open
Abstract
This study investigated whether radiomic features extracted from pretreatment [18F]FDG PET could improve the prediction of both histopathologic tumor response and survival in patients with locally advanced cervical cancer (LACC) treated with neoadjuvant chemoradiotherapy followed by surgery compared with conventional PET parameters and histopathologic features. Methods: The medical records of all consecutive patients with LACC referred between July 2010 and July 2016 were reviewed. [18F]FDG PET/CT was performed before neoadjuvant chemoradiotherapy. Radiomic features were extracted from the primary tumor volumes delineated semiautomatically on the PET images and reduced by factor analysis. A receiver-operating-characteristic analysis was performed, and conventional and radiomic features were dichotomized with Liu's method according to pathologic response (pR) and cancer-specific death. According to the study protocol, only areas under the curve of more than 0.70 were selected for further analysis, including logistic regression analysis for response prediction and Cox regression analysis for survival prediction. Results: A total of 195 patients fulfilled the inclusion criteria. At pathologic evaluation after surgery, 131 patients (67.2%) had no or microscopic (≤3 mm) residual tumor (pR0 or pR1, respectively); 64 patients (32.8%) had macroscopic residual tumor (>3 mm, pR2). With a median follow-up of 76.0 mo (95% CI, 70.7-78.7 mo), 31.3% of patients had recurrence or progression and 20.0% died of the disease. Among conventional PET parameters, SUVmean significantly differed between pathologic responders and nonresponders. Among radiomic features, 1 shape and 3 textural features significantly differed between pathologic responders and nonresponders. Three radiomic features significantly differed between presence and absence of recurrence or progression and between presence and absence of cancer-specific death. Areas under the curve were less than 0.70 for all parameters; thus, univariate and multivariate regression analyses were not performed. Conclusion: In a large series of patients with LACC treated with neoadjuvant chemoradiotherapy followed by surgery, PET radiomic features could not predict histopathologic tumor response and survival. It is crucial to further explore the biologic mechanism underlying imaging-derived parameters and plan a large, prospective, multicenter study with standardized protocols for all phases of the process of radiomic analysis to validate radiomics before its use in clinical routine.
Collapse
Affiliation(s)
- Angela Collarino
- Nuclear Medicine Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Vanessa Feudo
- Section of Nuclear Medicine, University Department of Radiological Sciences and Hematology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Tina Pasciuto
- Research Core Facility Data Collection G-STeP, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Anita Florit
- Section of Nuclear Medicine, University Department of Radiological Sciences and Hematology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Elisabeth Pfaehler
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Marco de Summa
- PET/CT Center, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Nicolò Bizzarri
- Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Salvatore Annunziata
- Nuclear Medicine Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Gian Franco Zannoni
- Gynecopathology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Section of Pathology, Department of Woman and Child Health and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lioe-Fee de Geus-Oei
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Biomedical Photonic Imaging Group, MIRA Institute, University of Twente, Enschede, The Netherlands
- Department of Radiation Science and Technology, Technical University of Delft, Delft, The Netherlands
| | - Gabriella Ferrandina
- Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Section of Obstetrics and Gynecology, University Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Maria Antonietta Gambacorta
- Radiation Oncology Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Roma, Italy
- Section of Radiology, University Department of Radiological Sciences and Hematology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Scambia
- Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Section of Obstetrics and Gynecology, University Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location VU University Medical Center, Amsterdam, The Netherlands; and
| | - Evis Sala
- Section of Radiology, University Department of Radiological Sciences and Hematology, Università Cattolica del Sacro Cuore, Rome, Italy
- Advanced Radiodiagnostics Centre, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Vittoria Rufini
- Nuclear Medicine Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy;
- Section of Nuclear Medicine, University Department of Radiological Sciences and Hematology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Floris Hp van Velden
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
5
|
Singh S, Mohajer B, Wells SA, Garg T, Hanneman K, Takahashi T, AlDandan O, McBee MP, Jawahar A. Imaging Genomics and Multiomics: A Guide for Beginners Starting Radiomics-Based Research. Acad Radiol 2024; 31:2281-2291. [PMID: 38286723 DOI: 10.1016/j.acra.2024.01.024] [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: 10/30/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/31/2024]
Abstract
Radiomics uses advanced mathematical analysis of pixel-level information from radiologic images to extract existing information in traditional imaging algorithms. It is intended to find imaging biomarkers related to the genomics of tumors or disease patterns that improve medical care by advanced detection of tumor response patterns in tumors and to assess prognosis. Radiomics expands the paradigm of medical imaging to help with diagnosis, management of diseases and prognostication, leveraging image features by extracting information that can be used as imaging biomarkers to predict prognosis and response to treatment. Radiogenomics is an emerging area in radiomics that investigates the association between imaging characteristics and gene expression profiles. There are an increasing number of research publications using different radiomics approaches without a clear consensus on which method works best. We aim to describe the workflow of radiomics along with a guide of what to expect when starting a radiomics-based research project.
Collapse
Affiliation(s)
- Shiva Singh
- Radiology and Imaging Sciences, National Institutes of Health, Bethesda, Maryland
| | - Bahram Mohajer
- Radiology and Radiological Sciences, Johns Hopkins Medicine, Baltimore, Maryland
| | - Shane A Wells
- Radiology, University of Michigan, Ann Arbor, Michigan
| | - Tushar Garg
- Radiology and Radiological Sciences, Johns Hopkins Medicine, Baltimore, Maryland
| | - Kate Hanneman
- Medical Imaging, University of Toronto, Toronto, ON, Canada
| | | | - Omran AlDandan
- Department of Radiology, Imam Abdulrahman Bin Faisal University, College of Medicine: Dammam, Eastern, Saudi Arabia
| | - Morgan P McBee
- Radiology and Radiological Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Anugayathri Jawahar
- Radiology, Northwestern University-Feinberg School of Medicine, 800, Arkes Pavilion, 676 N St. Clair St, Chicago, IL 60611.
| |
Collapse
|
6
|
Kocak B, Borgheresi A, Ponsiglione A, Andreychenko AE, Cavallo AU, Stanzione A, Doniselli FM, Vernuccio F, Triantafyllou M, Cannella R, Trotta R, Ghezzo S, Akinci D'Antonoli T, Cuocolo R. Explanation and Elaboration with Examples for CLEAR (CLEAR-E3): an EuSoMII Radiomics Auditing Group Initiative. Eur Radiol Exp 2024; 8:72. [PMID: 38740707 PMCID: PMC11091004 DOI: 10.1186/s41747-024-00471-z] [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: 03/18/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024] Open
Abstract
Overall quality of radiomics research has been reported as low in literature, which constitutes a major challenge to improve. Consistent, transparent, and accurate reporting is critical, which can be accomplished with systematic use of reporting guidelines. The CheckList for EvaluAtion of Radiomics research (CLEAR) was previously developed to assist authors in reporting their radiomic research and to assist reviewers in their evaluation. To take full advantage of CLEAR, further explanation and elaboration of each item, as well as literature examples, may be useful. The main goal of this work, Explanation and Elaboration with Examples for CLEAR (CLEAR-E3), is to improve CLEAR's usability and dissemination. In this international collaborative effort, members of the European Society of Medical Imaging Informatics-Radiomics Auditing Group searched radiomics literature to identify representative reporting examples for each CLEAR item. At least two examples, demonstrating optimal reporting, were presented for each item. All examples were selected from open-access articles, allowing users to easily consult the corresponding full-text articles. In addition to these, each CLEAR item's explanation was further expanded and elaborated. For easier access, the resulting document is available at https://radiomic.github.io/CLEAR-E3/ . As a complementary effort to CLEAR, we anticipate that this initiative will assist authors in reporting their radiomics research with greater ease and transparency, as well as editors and reviewers in reviewing manuscripts.Relevance statement Along with the original CLEAR checklist, CLEAR-E3 is expected to provide a more in-depth understanding of the CLEAR items, as well as concrete examples for reporting and evaluating radiomic research.Key points• As a complementary effort to CLEAR, this international collaborative effort aims to assist authors in reporting their radiomics research, as well as editors and reviewers in reviewing radiomics manuscripts.• Based on positive examples from the literature selected by the EuSoMII Radiomics Auditing Group, each CLEAR item explanation was further elaborated in CLEAR-E3.• The resulting explanation and elaboration document with examples can be accessed at https://radiomic.github.io/CLEAR-E3/ .
Collapse
Affiliation(s)
- Burak Kocak
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey.
| | - Alessandra Borgheresi
- Department of Clinical, Special and Dental Sciences, University Politecnica delle Marche, Ancona, Italy
- Department of Radiology, University Hospital "Azienda Ospedaliero Universitaria delle Marche", Via Conca 71, 60126, Ancona, Italy
| | - Andrea Ponsiglione
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Anna E Andreychenko
- Laboratory for Digital Public Health Technologies, ITMO University, St. Petersburg, Russian Federation
| | - Armando Ugo Cavallo
- Division of Radiology, Istituto Dermopatico dell'Immacolata (IDI) IRCCS, Rome, Italy
| | - Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Fabio M Doniselli
- Neuroradiology Unit, Fondazione Istituto Neurologico Carlo Besta, Via Celoria 11, 20133, Milano, Italy
| | - Federica Vernuccio
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnosis (Bi.N.D), University of Palermo, 90127, Palermo, Italy
| | - Matthaios Triantafyllou
- Department of Medical Imaging, University Hospital of Heraklion, 71110, Crete, Voutes, Greece
| | - Roberto Cannella
- Section of Radiology - Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Romina Trotta
- Department of Radiology - Fatima Hospital, Seville, Spain
| | | | - Tugba Akinci D'Antonoli
- Institute of Radiology and Nuclear Medicine, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Renato Cuocolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| |
Collapse
|
7
|
Hassan J, Saeed SM, Deka L, Uddin MJ, Das DB. Applications of Machine Learning (ML) and Mathematical Modeling (MM) in Healthcare with Special Focus on Cancer Prognosis and Anticancer Therapy: Current Status and Challenges. Pharmaceutics 2024; 16:260. [PMID: 38399314 PMCID: PMC10892549 DOI: 10.3390/pharmaceutics16020260] [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: 12/08/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
The use of data-driven high-throughput analytical techniques, which has given rise to computational oncology, is undisputed. The widespread use of machine learning (ML) and mathematical modeling (MM)-based techniques is widely acknowledged. These two approaches have fueled the advancement in cancer research and eventually led to the uptake of telemedicine in cancer care. For diagnostic, prognostic, and treatment purposes concerning different types of cancer research, vast databases of varied information with manifold dimensions are required, and indeed, all this information can only be managed by an automated system developed utilizing ML and MM. In addition, MM is being used to probe the relationship between the pharmacokinetics and pharmacodynamics (PK/PD interactions) of anti-cancer substances to improve cancer treatment, and also to refine the quality of existing treatment models by being incorporated at all steps of research and development related to cancer and in routine patient care. This review will serve as a consolidation of the advancement and benefits of ML and MM techniques with a special focus on the area of cancer prognosis and anticancer therapy, leading to the identification of challenges (data quantity, ethical consideration, and data privacy) which are yet to be fully addressed in current studies.
Collapse
Affiliation(s)
- Jasmin Hassan
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (S.M.S.)
| | | | - Lipika Deka
- Faculty of Computing, Engineering and Media, De Montfort University, Leicester LE1 9BH, UK;
| | - Md Jasim Uddin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Diganta B. Das
- Department of Chemical Engineering, Loughborough University, Loughborough LE11 3TU, UK
| |
Collapse
|
8
|
Demircioğlu A. The effect of data resampling methods in radiomics. Sci Rep 2024; 14:2858. [PMID: 38310165 PMCID: PMC10838284 DOI: 10.1038/s41598-024-53491-5] [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: 12/12/2023] [Accepted: 02/01/2024] [Indexed: 02/05/2024] Open
Abstract
Radiomic datasets can be class-imbalanced, for instance, when the prevalence of diseases varies notably, meaning that the number of positive samples is much smaller than that of negative samples. In these cases, the majority class may dominate the model's training and thus negatively affect the model's predictive performance, leading to bias. Therefore, resampling methods are often utilized to class-balance the data. However, several resampling methods exist, and neither their relative predictive performance nor their impact on feature selection has been systematically analyzed. In this study, we aimed to measure the impact of nine resampling methods on radiomic models utilizing a set of fifteen publicly available datasets regarding their predictive performance. Furthermore, we evaluated the agreement and similarity of the set of selected features. Our results show that applying resampling methods did not improve the predictive performance on average. On specific datasets, slight improvements in predictive performance (+ 0.015 in AUC) could be seen. A considerable disagreement on the set of selected features was seen (only 28.7% of features agreed), which strongly impedes feature interpretability. However, selected features are similar when considering their correlation (82.9% of features correlated on average).
Collapse
Affiliation(s)
- Aydin Demircioğlu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany.
| |
Collapse
|
9
|
Kocak B, Akinci D'Antonoli T, Mercaldo N, Alberich-Bayarri A, Baessler B, Ambrosini I, Andreychenko AE, Bakas S, Beets-Tan RGH, Bressem K, Buvat I, Cannella R, Cappellini LA, Cavallo AU, Chepelev LL, Chu LCH, Demircioglu A, deSouza NM, Dietzel M, Fanni SC, Fedorov A, Fournier LS, Giannini V, Girometti R, Groot Lipman KBW, Kalarakis G, Kelly BS, Klontzas ME, Koh DM, Kotter E, Lee HY, Maas M, Marti-Bonmati L, Müller H, Obuchowski N, Orlhac F, Papanikolaou N, Petrash E, Pfaehler E, Pinto Dos Santos D, Ponsiglione A, Sabater S, Sardanelli F, Seeböck P, Sijtsema NM, Stanzione A, Traverso A, Ugga L, Vallières M, van Dijk LV, van Griethuysen JJM, van Hamersvelt RW, van Ooijen P, Vernuccio F, Wang A, Williams S, Witowski J, Zhang Z, Zwanenburg A, Cuocolo R. METhodological RadiomICs Score (METRICS): a quality scoring tool for radiomics research endorsed by EuSoMII. Insights Imaging 2024; 15:8. [PMID: 38228979 PMCID: PMC10792137 DOI: 10.1186/s13244-023-01572-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: 08/01/2023] [Accepted: 11/20/2023] [Indexed: 01/18/2024] Open
Abstract
PURPOSE To propose a new quality scoring tool, METhodological RadiomICs Score (METRICS), to assess and improve research quality of radiomics studies. METHODS We conducted an online modified Delphi study with a group of international experts. It was performed in three consecutive stages: Stage#1, item preparation; Stage#2, panel discussion among EuSoMII Auditing Group members to identify the items to be voted; and Stage#3, four rounds of the modified Delphi exercise by panelists to determine the items eligible for the METRICS and their weights. The consensus threshold was 75%. Based on the median ranks derived from expert panel opinion and their rank-sum based conversion to importance scores, the category and item weights were calculated. RESULT In total, 59 panelists from 19 countries participated in selection and ranking of the items and categories. Final METRICS tool included 30 items within 9 categories. According to their weights, the categories were in descending order of importance: study design, imaging data, image processing and feature extraction, metrics and comparison, testing, feature processing, preparation for modeling, segmentation, and open science. A web application and a repository were developed to streamline the calculation of the METRICS score and to collect feedback from the radiomics community. CONCLUSION In this work, we developed a scoring tool for assessing the methodological quality of the radiomics research, with a large international panel and a modified Delphi protocol. With its conditional format to cover methodological variations, it provides a well-constructed framework for the key methodological concepts to assess the quality of radiomic research papers. CRITICAL RELEVANCE STATEMENT A quality assessment tool, METhodological RadiomICs Score (METRICS), is made available by a large group of international domain experts, with transparent methodology, aiming at evaluating and improving research quality in radiomics and machine learning. KEY POINTS • A methodological scoring tool, METRICS, was developed for assessing the quality of radiomics research, with a large international expert panel and a modified Delphi protocol. • The proposed scoring tool presents expert opinion-based importance weights of categories and items with a transparent methodology for the first time. • METRICS accounts for varying use cases, from handcrafted radiomics to entirely deep learning-based pipelines. • A web application has been developed to help with the calculation of the METRICS score ( https://metricsscore.github.io/metrics/METRICS.html ) and a repository created to collect feedback from the radiomics community ( https://github.com/metricsscore/metrics ).
Collapse
Affiliation(s)
- Burak Kocak
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Tugba Akinci D'Antonoli
- Institute of Radiology and Nuclear Medicine, Cantonal Hospital Baselland, Liestal, Switzerland.
| | - Nathaniel Mercaldo
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Bettina Baessler
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, Würzburg, Germany
| | - Ilaria Ambrosini
- Department of Translational Research, Academic Radiology, University of Pisa, Pisa, Italy
| | - Anna E Andreychenko
- Laboratory for Digital Public Health Technologies, ITMO University, St. Petersburg, Russian Federation
| | - Spyridon Bakas
- Division of Computational Pathology, Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN, USA
- Center for Federated Learning in Precision Medicine, Indiana University, Indianapolis, IN, USA
| | - Regina G H Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Keno Bressem
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Irene Buvat
- Institut Curie, Inserm, PSL University, Laboratory of Translational Imaging in Oncology, Orsay, France
| | - Roberto Cannella
- Section of Radiology - Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | | | - Armando Ugo Cavallo
- Division of Radiology, Istituto Dermopatico dell'Immacolata (IDI) IRCCS, Rome, Italy
| | - Leonid L Chepelev
- Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada
| | - Linda Chi Hang Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Aydin Demircioglu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital , Essen, Germany
| | - Nandita M deSouza
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
- Department of Imaging, The Royal Marsden National Health Service (NHS) Foundation Trust, London, UK
| | - Matthias Dietzel
- Department of Radiology, University Hospital Erlangen, Erlangen, Germany
| | | | - Andrey Fedorov
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Laure S Fournier
- Department of Radiology, Université Paris Cité, AP-HP, Hôpital Européen Georges Pompidou, PARCC UMRS 970, INSERM, Paris, France
| | | | - Rossano Girometti
- Institute of Radiology, Department of Medicine, University of Udine, University Hospital S. Maria della Misericordia, Udine, Italy
| | - Kevin B W Groot Lipman
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Georgios Kalarakis
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Science, Division of Radiology, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
- Department of Radiology, Medical School, University of Crete, Heraklion, Greece
| | - Brendan S Kelly
- Department of Radiology, St Vincent's University Hospital, Dublin, Ireland
- Insight Centre for Data Analytics, UCD, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Michail E Klontzas
- Department of Medical Imaging, University Hospital of Heraklion, Crete, Greece
- Department of Radiology, School of Medicine, University of Crete, Heraklion, Crete, Greece
- Computational Biomedicine Laboratory, Institute of Computer Science, FORTH, Heraklion, Crete, Greece
| | - Dow-Mu Koh
- Department of Radiology, Royal Marsden Hospital, Sutton, UK
| | - Elmar Kotter
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Ho Yun Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea
| | - Mario Maas
- Department of Radiology & Nuclear Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Luis Marti-Bonmati
- Medical Imaging Department and Biomedical Imaging Research Group, Hospital Universitario y Politécnico La Fe and Health Research Institute, Valencia, Spain
| | - Henning Müller
- University of Applied Sciences of Western Switzerland (HES-SO Valais), Sierra, Switzerland
- Department of Radiology and Medical Informatics, University of Geneva (UniGe), Geneva, Switzerland
| | - Nancy Obuchowski
- Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Fanny Orlhac
- Institut Curie, Inserm, PSL University, Laboratory of Translational Imaging in Oncology, Orsay, France
| | - Nikolaos Papanikolaou
- Computational Clinical Imaging Group, Centre for the Unknown, Champalimaud Foundation, Lisbon, Portugal
- Department of Radiology, Royal Marsden Hospital and The Institute of Cancer Research, London, UK
| | - Ekaterina Petrash
- Radiology department, Research Institute of Pediatric Oncology and Hematology n. a. L.A. Durnov, National Medical Research Center of Oncology n. a. N.N. Blokhin Ministry of Health of Russian Federation, Moscow, Russia
- Medical Department IRA-Labs, Moscow, Russia
| | - Elisabeth Pfaehler
- Institute for advanced simulation (IAS-8): Machine learning and data analytics, Forschungszentrum Jülich, Jülich, Germany
| | - Daniel Pinto Dos Santos
- Department of Radiology, University Hospital of Cologne, Cologne, Germany
- Institute for Diagnostic and Interventional Radiology, Goethe-University Frankfurt Am Main, Frankfurt, Germany
| | - Andrea Ponsiglione
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Sebastià Sabater
- Department of Radiation Oncology, Complejo Hospitalario Universitario de Albacete, Albacete, Spain
| | - Francesco Sardanelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
- Unit of Radiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Philipp Seeböck
- Computational Imaging Research Lab, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Nanna M Sijtsema
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Alberto Traverso
- Department of Radiotherapy, Maastro Clinic, Maastricht, the Netherlands
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Ugga
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Martin Vallières
- Department of Computer Science, Université de Sherbrooke, Sherbrooke, Canada
- Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, Canada
| | - Lisanne V van Dijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Robbert W van Hamersvelt
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Peter van Ooijen
- Department of Radiotherapy, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Federica Vernuccio
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnosis (Bi.N.D), University of Palermo, Palermo, 90127, Italy
| | - Alan Wang
- Centre for Medical Imaging & Centre for Brain Research, Faculty of Medical and Health Sciences, Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Stuart Williams
- Department of Radiology, Norfolk & Norwich University Hospital, Colney Lane, Norwich, Norfolk, UK
| | - Jan Witowski
- Department of Radiology, New York University Grossman School of Medicine, New York, USA
| | - Zhongyi Zhang
- School of Information and Communication Technology, Griffith University, Nathan, Brisbane, Australia
| | - Alex Zwanenburg
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Renato Cuocolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| |
Collapse
|
10
|
Martin A, Marcelin C, Petitpierre F, Jambon E, Maaloum R, Grenier N, Le Bras Y, Crombé A. Clinical, Technical, and MRI Features Associated with Patients' Outcome at 3 Months and 2 Years following Prostate Artery Embolization: Is There an Added Value of Radiomics? J Pers Med 2024; 14:67. [PMID: 38248768 PMCID: PMC10817287 DOI: 10.3390/jpm14010067] [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: 10/20/2023] [Revised: 12/26/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
Our aim was to investigate which features were associated with clinical successes at short- and mid-terms following prostate artery embolization (PAE) for symptomatic benign prostate hypertrophy (BPH). All adults treated by PAE for BPH at our referral center between January 2017 and March 2021, with pre-treatment MRI, technical success, and follow-up at 3 months and 2 years were included in this single-center retrospective study. Radiologists reviewed the prostatic protrusion index (PPI), adenomatous dominant BPH (adBPH), and Wasserman classification on pre-treatment MRI. Radiomics analysis was achieved on the transitional zone on pre-treatment T2-weighted imaging (WI) and ADC, and comprised reproducibility assessment, unsupervised classifications, and supervised radiomics scores obtained with cross-validated Elasticnet regressions. Eighty-eight patients were included (median age: 65 years), with 81.8% clinical successes at 3 months and 60.2% at 2 years. No feature was associated with success at 3 months, except the radiomics score trained on T2-WI and ADC (AUROC = 0.694). Regarding success at 2 years, no radiomics approaches provided significant performances; however, Wasserman type-1 and change in international prostate symptom score (IPSS) at 3 months ≤ -35% were associated with success in multivariable analysis (OR = 5.82, p = 0.0296, and OR = 9.04, p = 0.0002). Thus, while radiomics provided limited interest, Wasserman classification and early IPSS changes appeared predictive of mid-term outcomes.
Collapse
Affiliation(s)
- Antoine Martin
- Department of Diagnostic and Interventional Radiology, Pellegrin University Hospital, Place Amélie-Raba-Léon, F-33076 Bordeaux, France (F.P.); (R.M.); (N.G.)
| | - Clément Marcelin
- Department of Diagnostic and Interventional Radiology, Pellegrin University Hospital, Place Amélie-Raba-Léon, F-33076 Bordeaux, France (F.P.); (R.M.); (N.G.)
- BRIC Bordeaux Institute of Oncology, INSERM U1312, 2 Rue Dr Hoffmann Martinot, F-33000 Bordeaux, France
| | - François Petitpierre
- Department of Diagnostic and Interventional Radiology, Pellegrin University Hospital, Place Amélie-Raba-Léon, F-33076 Bordeaux, France (F.P.); (R.M.); (N.G.)
- Department of Radiology, Clinique Mutualiste de Pessac, 46 Avenue du Dr Albert Schweitzer, F-33600 Pessac, France
| | - Eva Jambon
- Department of Diagnostic and Interventional Radiology, Pellegrin University Hospital, Place Amélie-Raba-Léon, F-33076 Bordeaux, France (F.P.); (R.M.); (N.G.)
| | - Rim Maaloum
- Department of Diagnostic and Interventional Radiology, Pellegrin University Hospital, Place Amélie-Raba-Léon, F-33076 Bordeaux, France (F.P.); (R.M.); (N.G.)
| | - Nicolas Grenier
- Department of Diagnostic and Interventional Radiology, Pellegrin University Hospital, Place Amélie-Raba-Léon, F-33076 Bordeaux, France (F.P.); (R.M.); (N.G.)
| | - Yann Le Bras
- Department of Diagnostic and Interventional Radiology, Pellegrin University Hospital, Place Amélie-Raba-Léon, F-33076 Bordeaux, France (F.P.); (R.M.); (N.G.)
| | - Amandine Crombé
- Department of Diagnostic and Interventional Radiology, Pellegrin University Hospital, Place Amélie-Raba-Léon, F-33076 Bordeaux, France (F.P.); (R.M.); (N.G.)
- BRIC Bordeaux Institute of Oncology, INSERM U1312, 2 Rue Dr Hoffmann Martinot, F-33000 Bordeaux, France
| |
Collapse
|
11
|
Huang W, Tao Z, Younis MH, Cai W, Kang L. Nuclear medicine radiomics in digestive system tumors: Concept, applications, challenges, and future perspectives. VIEW 2023; 4:20230032. [PMID: 38179181 PMCID: PMC10766416 DOI: 10.1002/viw.20230032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/20/2023] [Indexed: 01/06/2024] Open
Abstract
Radiomics aims to develop novel biomarkers and provide relevant deeper subvisual information about pathology, immunophenotype, and tumor microenvironment. It uses automated or semiautomated quantitative analysis of high-dimensional images to improve characterization, diagnosis, and prognosis. Recent years have seen a rapid increase in radiomics applications in nuclear medicine, leading to some promising research results in digestive system oncology, which have been driven by big data analysis and the development of artificial intelligence. Although radiomics advances one step further toward the non-invasive precision medical analysis, it is still a step away from clinical application and faces many challenges. This review article summarizes the available literature on digestive system tumors regarding radiomics in nuclear medicine. First, we describe the workflow and steps involved in radiomics analysis. Subsequently, we discuss the progress in clinical application regarding the utilization of radiomics for distinguishing between various diseases and evaluating their prognosis, and demonstrate how radiomics advances this field. Finally, we offer our viewpoint on how the field can progress by addressing the challenges facing clinical implementation.
Collapse
Affiliation(s)
- Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Zihao Tao
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Muhsin H. Younis
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| |
Collapse
|
12
|
Fuchs T, Kaiser L, Müller D, Papp L, Fischer R, Tran-Gia J. Enhancing Interoperability and Harmonisation of Nuclear Medicine Image Data and Associated Clinical Data. Nuklearmedizin 2023; 62:389-398. [PMID: 37907246 PMCID: PMC10689089 DOI: 10.1055/a-2187-5701] [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: 09/05/2023] [Accepted: 09/21/2023] [Indexed: 11/02/2023]
Abstract
Nuclear imaging techniques such as positron emission tomography (PET) and single photon emission computed tomography (SPECT) in combination with computed tomography (CT) are established imaging modalities in clinical practice, particularly for oncological problems. Due to a multitude of manufacturers, different measurement protocols, local demographic or clinical workflow variations as well as various available reconstruction and analysis software, very heterogeneous datasets are generated. This review article examines the current state of interoperability and harmonisation of image data and related clinical data in the field of nuclear medicine. Various approaches and standards to improve data compatibility and integration are discussed. These include, for example, structured clinical history, standardisation of image acquisition and reconstruction as well as standardised preparation of image data for evaluation. Approaches to improve data acquisition, storage and analysis will be presented. Furthermore, approaches are presented to prepare the datasets in such a way that they become usable for projects applying artificial intelligence (AI) (machine learning, deep learning, etc.). This review article concludes with an outlook on future developments and trends related to AI in nuclear medicine, including a brief research of commercial solutions.
Collapse
Affiliation(s)
- Timo Fuchs
- Medical Data Integration Center (MEDIZUKR), University Hospital Regensburg, Regensburg, Germany
- Partner Site Regensburg, Bavarian Center for Cancer Research (BZKF), Regensburg, Germany
| | - Lena Kaiser
- Department of Nuclear Medicine, LMU University Hospital, LMU, Munich, Germany
| | - Dominik Müller
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
- Medical Data Integration Center, University Hospital Augsburg, Augsburg, Germany
| | - Laszlo Papp
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Wien, Austria
| | - Regina Fischer
- Medical Data Integration Center (MEDIZUKR), University Hospital Regensburg, Regensburg, Germany
- Partner Site Regensburg, Bavarian Center for Cancer Research (BZKF), Regensburg, Germany
| | - Johannes Tran-Gia
- Department of Nuclear Medicine, University Hospital Würzburg, Wurzburg, Germany
| |
Collapse
|
13
|
Luining WI, Oprea-Lager DE, Vis AN, van Moorselaar RJA, Knol RJJ, Wondergem M, Boellaard R, Cysouw MCF. Optimization and validation of 18F-DCFPyL PET radiomics-based machine learning models in intermediate- to high-risk primary prostate cancer. PLoS One 2023; 18:e0293672. [PMID: 37943772 PMCID: PMC10635444 DOI: 10.1371/journal.pone.0293672] [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: 04/25/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023] Open
Abstract
INTRODUCTION Radiomics extracted from prostate-specific membrane antigen (PSMA)-PET modeled with machine learning (ML) may be used for prediction of disease risk. However, validation of previously proposed approaches is lacking. We aimed to optimize and validate ML models based on 18F-DCFPyL-PET radiomics for the prediction of lymph-node involvement (LNI), extracapsular extension (ECE), and postoperative Gleason score (GS) in primary prostate cancer (PCa) patients. METHODS Patients with intermediate- to high-risk PCa who underwent 18F-DCFPyL-PET/CT before radical prostatectomy with pelvic lymph-node dissection were evaluated. The training dataset included 72 patients, the internal validation dataset 24 patients, and the external validation dataset 27 patients. PSMA-avid intra-prostatic lesions were delineated semi-automatically on PET and 480 radiomics features were extracted. Conventional PET-metrics were derived for comparative analysis. Segmentation, preprocessing, and ML methods were optimized in repeated 5-fold cross-validation (CV) on the training dataset. The trained models were tested on the combined validation dataset. Combat harmonization was applied to external radiomics data. Model performance was assessed using the receiver-operating-characteristics curve (AUC). RESULTS The CV-AUCs in the training dataset were 0.88, 0.79 and 0.84 for LNI, ECE, and GS, respectively. In the combined validation dataset, the ML models could significantly predict GS with an AUC of 0.78 (p<0.05). However, validation AUCs for LNI and ECE prediction were not significant (0.57 and 0.63, respectively). Conventional PET metrics-based models had comparable AUCs for LNI (0.59, p>0.05) and ECE (0.66, p>0.05), but a lower AUC for GS (0.73, p<0.05). In general, Combat harmonization improved external validation AUCs (-0.03 to +0.18). CONCLUSION In internal and external validation, 18F-DCFPyL-PET radiomics-based ML models predicted high postoperative GS but not LNI or ECE in intermediate- to high-risk PCa. Therefore, the clinical benefit seems to be limited. These results underline the need for external and/or multicenter validation of PET radiomics-based ML model analyses to assess their generalizability.
Collapse
Affiliation(s)
- Wietske I. Luining
- Department of Urology, Amsterdam University Medical Centers, Prostate Cancer Network Netherlands, Amsterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Daniela E. Oprea-Lager
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - André N. Vis
- Department of Urology, Amsterdam University Medical Centers, Prostate Cancer Network Netherlands, Amsterdam, The Netherlands
| | - Reindert J. A. van Moorselaar
- Department of Urology, Amsterdam University Medical Centers, Prostate Cancer Network Netherlands, Amsterdam, The Netherlands
| | - Remco J. J. Knol
- Department of Nuclear Medicine, Northwest Clinics, Alkmaar, The Netherlands
| | - Maurits Wondergem
- Department of Nuclear Medicine, Northwest Clinics, Alkmaar, The Netherlands
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Matthijs C. F. Cysouw
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Kocak B, Yuzkan S, Mutlu S, Bulut E, Kavukoglu I. Publications poorly report the essential RadiOmics ParametERs (PROPER): A meta-research on quality of reporting. Eur J Radiol 2023; 167:111088. [PMID: 37713968 DOI: 10.1016/j.ejrad.2023.111088] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/15/2023] [Accepted: 09/05/2023] [Indexed: 09/17/2023]
Abstract
PURPOSE To investigate the quality of reporting in radiomics research, with a focus on the most basic technical parameters. METHODS A PubMed literature search was conducted to identify original studies on radiomics (last search: January 2, 2023). Following a sample size calculation with an a priori power analysis, a random sample of the radiomic literature was collected. In addition to baseline characteristics, the key aspects of radiomic software, resampling, and discretization were evaluated. Agreement between raters was analyzed. Disagreements were resolved through consensus. RESULTS A sample of 87 publications was evaluated. Most publications (89%; 77/87) were retrospective. They were conducted predominantly with private data (87%; 76/87) at a single institution (77%; 67/87) without external validation (90%; 78/87). 69% (60/87) of the papers reported the radiomic software used (p < 0.001), with nearly half (43%; 26/60) omitting the version. 37% (32/87) reported the resampling size (p = 0.018), while 22% (7/32) did not report using iso-voxel resampling. 34% (30/87) reported the discretization parameters (p < 0.01), but more than three-quarters (77%; 23/30) did not experiment with different discretization parameters. A wide range of discretization parameter values were reported. Most papers (79%; 69/87) failed to report all three essential items simultaneously (p < 0.001). CONCLUSION Even the essential radiomic parameters that are usually displayed on the user interface of radiomic software tools were poorly reported in radiomics-related publications. This issue of transparency may require additional action from researchers, editors, and reviewers in the form of adopting more stringent reporting standards (e.g., checklists, guidelines).
Collapse
Affiliation(s)
- Burak Kocak
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey.
| | - Sabahattin Yuzkan
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Samet Mutlu
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Elif Bulut
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Irem Kavukoglu
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| |
Collapse
|
15
|
Ahmed TM, Kawamoto S, Hruban RH, Fishman EK, Soyer P, Chu LC. A primer on artificial intelligence in pancreatic imaging. Diagn Interv Imaging 2023; 104:435-447. [PMID: 36967355 DOI: 10.1016/j.diii.2023.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 06/18/2023]
Abstract
Artificial Intelligence (AI) is set to transform medical imaging by leveraging the vast data contained in medical images. Deep learning and radiomics are the two main AI methods currently being applied within radiology. Deep learning uses a layered set of self-correcting algorithms to develop a mathematical model that best fits the data. Radiomics converts imaging data into mineable features such as signal intensity, shape, texture, and higher-order features. Both methods have the potential to improve disease detection, characterization, and prognostication. This article reviews the current status of artificial intelligence in pancreatic imaging and critically appraises the quality of existing evidence using the radiomics quality score.
Collapse
Affiliation(s)
- Taha M Ahmed
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ralph H Hruban
- Sol Goldman Pancreatic Research Center, Department of Pathology, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Philippe Soyer
- Université Paris Cité, Faculté de Médecine, Department of Radiology, Hôpital Cochin-APHP, 75014, 75006, Paris, France, 7501475006
| | - Linda C Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
16
|
Pinto Dos Santos D. A meta-discussion on radiomics - Meta-research, bias, quality and other issues. Eur J Radiol 2023; 166:111007. [PMID: 37531696 DOI: 10.1016/j.ejrad.2023.111007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/04/2023]
Affiliation(s)
- Daniel Pinto Dos Santos
- Department of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany; Department of Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| |
Collapse
|
17
|
Mohebi M, Amini M, Alemzadeh-Ansari MJ, Alizadehasl A, Rajabi AB, Shiri I, Zaidi H, Orooji M. Post-revascularization Ejection Fraction Prediction for Patients Undergoing Percutaneous Coronary Intervention Based on Myocardial Perfusion SPECT Imaging Radiomics: a Preliminary Machine Learning Study. J Digit Imaging 2023; 36:1348-1363. [PMID: 37059890 PMCID: PMC10407007 DOI: 10.1007/s10278-023-00820-1] [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: 09/21/2022] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/16/2023] Open
Abstract
In this study, the ability of radiomics features extracted from myocardial perfusion imaging with SPECT (MPI-SPECT) was investigated for the prediction of ejection fraction (EF) post-percutaneous coronary intervention (PCI) treatment. A total of 52 patients who had undergone pre-PCI MPI-SPECT were enrolled in this study. After normalization of the images, features were extracted from the left ventricle, initially automatically segmented by k-means and active contour methods, and finally edited and approved by an expert radiologist. More than 1700 2D and 3D radiomics features were extracted from each patient's scan. A cross-combination of three feature selections and seven classifier methods was implemented. Three classes of no or dis-improvement (class 1), improved EF from 0 to 5% (class 2), and improved EF over 5% (class 3) were predicted by using tenfold cross-validation. Lastly, the models were evaluated based on accuracy, AUC, sensitivity, specificity, precision, and F-score. Neighborhood component analysis (NCA) selected the most predictive feature signatures, including Gabor, first-order, and NGTDM features. Among the classifiers, the best performance was achieved by the fine KNN classifier, which yielded mean accuracy, AUC, sensitivity, specificity, precision, and F-score of 0.84, 0.83, 0.75, 0.87, 0.78, and 0.76, respectively, in 100 iterations of classification, within the 52 patients with 10-fold cross-validation. The MPI-SPECT-based radiomic features are well suited for predicting post-revascularization EF and therefore provide a helpful approach for deciding on the most appropriate treatment.
Collapse
Affiliation(s)
- Mobin Mohebi
- Department of Biomedical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Amini
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva 4, Switzerland
| | | | - Azin Alizadehasl
- Echocardiography Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
- Cardio-Oncology Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Bitarafan Rajabi
- Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
- Echocardiography Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva 4, Switzerland
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva 4, Switzerland
- Geneva University Neuro Center, Geneva University, Geneva, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark
| | - Mahdi Orooji
- Department of Biomedical Engineering, Tarbiat Modares University, Tehran, Iran
- Department of Electrical and Computer Engineering, University of California–Davis, Davis, CA USA
| |
Collapse
|
18
|
Stanzione A, Cuocolo R, Bombace C, Pesce I, Mainolfi CG, De Giorgi M, Delli Paoli G, La Selva P, Petrone J, Camera L, Klain M, Del Vecchio S, Cuocolo A, Maurea S. Prediction of 2-[ 18F]FDG PET-CT SUVmax for Adrenal Mass Characterization: A CT Radiomics Feasibility Study. Cancers (Basel) 2023; 15:3439. [PMID: 37444549 DOI: 10.3390/cancers15133439] [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: 04/17/2023] [Revised: 06/20/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Indeterminate adrenal masses (AM) pose a diagnostic challenge, and 2-[18F]FDG PET-CT serves as a problem-solving tool. Aim of this study was to investigate whether CT radiomics features could be used to predict the 2-[18F]FDG SUVmax of AM. METHODS Patients with AM on 2-[18F]FDG PET-CT scan were grouped based on iodine contrast injection as CT contrast-enhanced (CE) or CT unenhanced (NCE). Two-dimensional segmentations of AM were manually obtained by multiple operators on CT images. Image resampling and discretization (bin number = 16) were performed. 919 features were calculated using PyRadiomics. After scaling, unstable, redundant, and low variance features were discarded. Using linear regression and the Uniform Manifold Approximation and Projection technique, a CT radiomics synthetic value (RadSV) was obtained. The correlation between CT RadSV and 2-[18F]FDG SUVmax was assessed with Pearson test. RESULTS A total of 725 patients underwent PET-CT from April 2020 to April 2021. In 150 (21%) patients, a total of 179 AM (29 bilateral) were detected. Group CE consisted of 84 patients with 108 AM (size = 18.1 ± 4.9 mm) and Group NCE of 66 patients with 71 AM (size = 18.5 ± 3.8 mm). In both groups, 39 features were selected. No statisticallyf significant correlation between CT RadSV and 2-[18F]FDG SUVmax was found (Group CE, r = 0.18 and p = 0.058; Group NCE, r = 0.13 and p = 0.27). CONCLUSIONS It might not be feasible to predict 2-[18F]FDG SUVmax of AM using CT RadSV. Its role as a problem-solving tool for indeterminate AM remains fundamental.
Collapse
Affiliation(s)
- Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Renato Cuocolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84084 Baronissi, Italy
| | - Claudia Bombace
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Ilaria Pesce
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Ciro Gabriele Mainolfi
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Marco De Giorgi
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Gregorio Delli Paoli
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Pasquale La Selva
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Jessica Petrone
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Luigi Camera
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Michele Klain
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Silvana Del Vecchio
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Simone Maurea
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| |
Collapse
|
19
|
Pullen LCE, Noortman WA, Triemstra L, de Jongh C, Rademaker FJ, Spijkerman R, Kalisvaart GM, Gertsen EC, de Geus-Oei LF, Tolboom N, de Steur WO, Dantuma M, Slart RHJA, van Hillegersberg R, Siersema PD, Ruurda JP, van Velden FHP, Vegt E. Prognostic Value of [ 18F]FDG PET Radiomics to Detect Peritoneal and Distant Metastases in Locally Advanced Gastric Cancer-A Side Study of the Prospective Multicentre PLASTIC Study. Cancers (Basel) 2023; 15:cancers15112874. [PMID: 37296837 DOI: 10.3390/cancers15112874] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/13/2023] [Accepted: 05/14/2023] [Indexed: 06/12/2023] Open
Abstract
AIM To improve identification of peritoneal and distant metastases in locally advanced gastric cancer using [18F]FDG-PET radiomics. METHODS [18F]FDG-PET scans of 206 patients acquired in 16 different Dutch hospitals in the prospective multicentre PLASTIC-study were analysed. Tumours were delineated and 105 radiomic features were extracted. Three classification models were developed to identify peritoneal and distant metastases (incidence: 21%): a model with clinical variables, a model with radiomic features, and a clinicoradiomic model, combining clinical variables and radiomic features. A least absolute shrinkage and selection operator (LASSO) regression classifier was trained and evaluated in a 100-times repeated random split, stratified for the presence of peritoneal and distant metastases. To exclude features with high mutual correlations, redundancy filtering of the Pearson correlation matrix was performed (r = 0.9). Model performances were expressed by the area under the receiver operating characteristic curve (AUC). In addition, subgroup analyses based on Lauren classification were performed. RESULTS None of the models could identify metastases with low AUCs of 0.59, 0.51, and 0.56, for the clinical, radiomic, and clinicoradiomic model, respectively. Subgroup analysis of intestinal and mixed-type tumours resulted in low AUCs of 0.67 and 0.60 for the clinical and radiomic models, and a moderate AUC of 0.71 in the clinicoradiomic model. Subgroup analysis of diffuse-type tumours did not improve the classification performance. CONCLUSION Overall, [18F]FDG-PET-based radiomics did not contribute to the preoperative identification of peritoneal and distant metastases in patients with locally advanced gastric carcinoma. In intestinal and mixed-type tumours, the classification performance of the clinical model slightly improved with the addition of radiomic features, but this slight improvement does not outweigh the laborious radiomic analysis.
Collapse
Affiliation(s)
- Lieke C E Pullen
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
| | - Wyanne A Noortman
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
- Department of Radiology, Leiden University Medical Center, 2333 ZD Leiden, The Netherlands
| | - Lianne Triemstra
- Department of Surgery, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Cas de Jongh
- Department of Surgery, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Fenna J Rademaker
- TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Romy Spijkerman
- TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Gijsbert M Kalisvaart
- Department of Radiology, Leiden University Medical Center, 2333 ZD Leiden, The Netherlands
| | - Emma C Gertsen
- Department of Surgery, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
- Department of Radiology, Leiden University Medical Center, 2333 ZD Leiden, The Netherlands
| | - Nelleke Tolboom
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Wobbe O de Steur
- Department of Surgery, Leiden University Medical Center, 2333 ZD Leiden, The Netherlands
| | - Maura Dantuma
- Multi-Modality Medical Imaging Group, TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Riemer H J A Slart
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | | | - Peter D Siersema
- Department of Gastroenterology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Jelle P Ruurda
- Department of Surgery, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Floris H P van Velden
- Department of Radiology, Leiden University Medical Center, 2333 ZD Leiden, The Netherlands
| | - Erik Vegt
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
20
|
Noortman WA, Aide N, Vriens D, Arkes LS, Slump CH, Boellaard R, Goeman JJ, Deroose CM, Machiels JP, Licitra LF, Lhommel R, Alessi A, Woff E, Goffin K, Le Tourneau C, Gal J, Temam S, Delord JP, van Velden FHP, de Geus-Oei LF. Development and External Validation of a PET Radiomic Model for Prognostication of Head and Neck Cancer. Cancers (Basel) 2023; 15:2681. [PMID: 37345017 DOI: 10.3390/cancers15102681] [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: 01/31/2023] [Revised: 04/30/2023] [Accepted: 05/03/2023] [Indexed: 06/23/2023] Open
Abstract
AIM To build and externally validate an [18F]FDG PET radiomic model to predict overall survival in patients with head and neck squamous cell carcinoma (HNSCC). METHODS Two multicentre datasets of patients with operable HNSCC treated with preoperative afatinib who underwent a baseline and evaluation [18F]FDG PET/CT scan were included (EORTC: n = 20, Unicancer: n = 34). Tumours were delineated, and radiomic features were extracted. Each cohort served once as a training and once as an external validation set for the prediction of overall survival. Supervised feature selection was performed using variable hunting with variable importance, selecting the top two features. A Cox proportional hazards regression model using selected radiomic features and clinical characteristics was fitted on the training dataset and validated in the external validation set. Model performances are expressed by the concordance index (C-index). RESULTS In both models, the radiomic model surpassed the clinical model with validation C-indices of 0.69 and 0.79 vs. 0.60 and 0.67, respectively. The model that combined the radiomic features and clinical variables performed best, with validation C-indices of 0.71 and 0.82. CONCLUSION Although assessed in two small but independent cohorts, an [18F]FDG-PET radiomic signature based on the evaluation scan seems promising for the prediction of overall survival for HNSSC treated with preoperative afatinib. The robustness and clinical applicability of this radiomic signature should be assessed in a larger cohort.
Collapse
Affiliation(s)
- Wyanne A Noortman
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Nicolas Aide
- Nuclear Medicine Department, Centre Hospitalier Universitaire de Caen, 14000 Caen, France
| | - Dennis Vriens
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Lisa S Arkes
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Technical Medicine, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Cornelis H Slump
- TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Ronald Boellaard
- Amsterdam University Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Jelle J Goeman
- Department of Biomedical Data Sciences, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Christophe M Deroose
- Nuclear Medicine and Molecular Imaging, Department of Imaging & Pathology, University Hospitals Leuven, KU Leuven, 3000 Leuven, Belgium
| | - Jean-Pascal Machiels
- Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Institute for Experimental and Clinical Research (IREC, pôle MIRO), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Lisa F Licitra
- Department of Head and Neck Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, University of Milan, 20133 Milan, Italy
| | - Renaud Lhommel
- Division of Nuclear Medicine, Institut de Recherche Clinique, Cliniques Universitaires Saint Luc, 1200 Brussels, Belgium
| | - Alessandra Alessi
- Department of Nuclear Medicine-PET Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Erwin Woff
- Nuclear Medicine Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B.), 1070 Bruxelles, Belgium
| | - Karolien Goffin
- Nuclear Medicine and Molecular Imaging, Department of Imaging & Pathology, University Hospitals Leuven, KU Leuven, 3000 Leuven, Belgium
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation, Institut Curie, Paris-Saclay University, 75005 Paris, France
| | - Jocelyn Gal
- Epidemiology and Biostatistics Department, Centre Antoine Lacassagne, University Côte d'Azur, 06100 Nice, France
| | - Stéphane Temam
- Department of Head and Neck Surgery Gustave Roussy, 94805 Villejuif, France
| | | | - Floris H P van Velden
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
- Department of Radiation Science & Technology, Delft University of Technology, 2628 CD Delft, The Netherlands
| |
Collapse
|
21
|
Kocak B, Baessler B, Bakas S, Cuocolo R, Fedorov A, Maier-Hein L, Mercaldo N, Müller H, Orlhac F, Pinto Dos Santos D, Stanzione A, Ugga L, Zwanenburg A. CheckList for EvaluAtion of Radiomics research (CLEAR): a step-by-step reporting guideline for authors and reviewers endorsed by ESR and EuSoMII. Insights Imaging 2023; 14:75. [PMID: 37142815 PMCID: PMC10160267 DOI: 10.1186/s13244-023-01415-8] [Citation(s) in RCA: 116] [Impact Index Per Article: 116.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/24/2023] [Indexed: 05/06/2023] Open
Abstract
Even though radiomics can hold great potential for supporting clinical decision-making, its current use is mostly limited to academic research, without applications in routine clinical practice. The workflow of radiomics is complex due to several methodological steps and nuances, which often leads to inadequate reporting and evaluation, and poor reproducibility. Available reporting guidelines and checklists for artificial intelligence and predictive modeling include relevant good practices, but they are not tailored to radiomic research. There is a clear need for a complete radiomics checklist for study planning, manuscript writing, and evaluation during the review process to facilitate the repeatability and reproducibility of studies. We here present a documentation standard for radiomic research that can guide authors and reviewers. Our motivation is to improve the quality and reliability and, in turn, the reproducibility of radiomic research. We name the checklist CLEAR (CheckList for EvaluAtion of Radiomics research), to convey the idea of being more transparent. With its 58 items, the CLEAR checklist should be considered a standardization tool providing the minimum requirements for presenting clinical radiomics research. In addition to a dynamic online version of the checklist, a public repository has also been set up to allow the radiomics community to comment on the checklist items and adapt the checklist for future versions. Prepared and revised by an international group of experts using a modified Delphi method, we hope the CLEAR checklist will serve well as a single and complete scientific documentation tool for authors and reviewers to improve the radiomics literature.
Collapse
Affiliation(s)
- Burak Kocak
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, 34480, Turkey.
| | - Bettina Baessler
- Institute of Diagnostic and Interventional Radiology, University Hospital Würzburg, Würzburg, Germany
| | - Spyridon Bakas
- Center for Artificial Intelligence for Integrated Diagnostics (AI2D) & Center for Biomedical Image Computing & Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Renato Cuocolo
- Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi, Italy
| | - Andrey Fedorov
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lena Maier-Hein
- Division of Intelligent Medical Systems, German Cancer Research Center, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Nathaniel Mercaldo
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Henning Müller
- University of Applied Sciences of Western Switzerland (HES-SO Valais), Valais, Switzerland
- Department of Radiology and Medical Informatics, University of Geneva (UniGe), Geneva, Switzerland
| | - Fanny Orlhac
- Laboratoire d'Imagerie Translationnelle en Oncologie (LITO)-U1288, Institut Curie, Inserm, Université PSL, Orsay, France
| | - Daniel Pinto Dos Santos
- Department of Radiology, University Hospital of Cologne, Cologne, Germany
- Institute for Diagnostic and Interventional Radiology, Goethe-University Frankfurt Am Main, Frankfurt, Germany
| | - Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Lorenzo Ugga
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Alex Zwanenburg
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
22
|
Kocak B, Bulut E, Bayrak ON, Okumus AA, Altun O, Borekci Arvas Z, Kavukoglu I. NEgatiVE results in Radiomics research (NEVER): A meta-research study of publication bias in leading radiology journals. Eur J Radiol 2023; 163:110830. [PMID: 37119709 DOI: 10.1016/j.ejrad.2023.110830] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/01/2023]
Abstract
PURPOSE The purpose of this study was to conduct a meta-research of radiomics-related articles for the publication of negative results, with a focus on the leading clinical radiology journals due to their purportedly high editorial standards. METHODS A literature search was performed in PubMed to identify original research studies on radiomics (last search date: August 16th, 2022). The search was restricted to studies published in Q1 clinical radiology journals indexed by Scopus and Web of Science. Following an a priori power analysis based on our null hypothesis, a random sampling of the published literature was conducted. Besides the six baseline study characteristics, a total of three items about publication bias were evaluated. Agreement between raters was analyzed. Disagreements were resolved through consensus. Statistical synthesis of the qualitative evaluations was presented. RESULTS Following a priori power analysis, we included a random sample of 149 publications in this study. Most of the publications were retrospective (95%; 142/149), based on private data (91%; 136/149), centered on a single institution (75%; 111/149), and lacked external validation (81%; 121/149). Slightly fewer than half (44%; 66/149) made no comparison to non-radiomic approaches. Overall, only one study (1%; 1/149) reported negative results for radiomics, yielding a statistically significant binomial test (p < 0.0001). CONCLUSION The top clinical radiology journals almost never publish negative results, having a strong bias toward publishing positive results. Almost half of the publications did not even compare their approach with a non-radiomic method.
Collapse
Affiliation(s)
- Burak Kocak
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey.
| | - Elif Bulut
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Osman Nuri Bayrak
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Ahmet Arda Okumus
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Omer Altun
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Zeynep Borekci Arvas
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Irem Kavukoglu
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| |
Collapse
|
23
|
Stanzione A, Ponsiglione A, Alessandrino F, Brembilla G, Imbriaco M. Beyond diagnosis: is there a role for radiomics in prostate cancer management? Eur Radiol Exp 2023; 7:13. [PMID: 36907973 PMCID: PMC10008761 DOI: 10.1186/s41747-023-00321-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/05/2023] [Indexed: 03/13/2023] Open
Abstract
The role of imaging in pretreatment staging and management of prostate cancer (PCa) is constantly evolving. In the last decade, there has been an ever-growing interest in radiomics as an image analysis approach able to extract objective quantitative features that are missed by human eye. However, most of PCa radiomics studies have been focused on cancer detection and characterisation. With this narrative review we aimed to provide a synopsis of the recently proposed potential applications of radiomics for PCa with a management-based approach, focusing on primary treatments with curative intent and active surveillance as well as highlighting on recurrent disease after primary treatment. Current evidence is encouraging, with radiomics and artificial intelligence appearing as feasible tools to aid physicians in planning PCa management. However, the lack of external independent datasets for validation and prospectively designed studies casts a shadow on the reliability and generalisability of radiomics models, delaying their translation into clinical practice.Key points• Artificial intelligence solutions have been proposed to streamline prostate cancer radiotherapy planning.• Radiomics models could improve risk assessment for radical prostatectomy patient selection.• Delta-radiomics appears promising for the management of patients under active surveillance.• Radiomics might outperform current nomograms for prostate cancer recurrence risk assessment.• Reproducibility of results, methodological and ethical issues must still be faced before clinical implementation.
Collapse
Affiliation(s)
- Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Andrea Ponsiglione
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy.
| | | | - Giorgio Brembilla
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Imbriaco
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
24
|
Prognostic analysis of curatively resected pancreatic cancer using harmonized positron emission tomography radiomic features. Eur J Hybrid Imaging 2023; 7:5. [PMID: 36872413 PMCID: PMC9986192 DOI: 10.1186/s41824-023-00163-8] [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: 12/05/2022] [Accepted: 01/18/2023] [Indexed: 03/07/2023] Open
Abstract
BACKGROUND Texture features reflecting tumour heterogeneity enable us to investigate prognostic factors. The R package ComBat can harmonize the quantitative texture features among several positron emission tomography (PET) scanners. We aimed to identify prognostic factors among harmonized PET radiomic features and clinical information from pancreatic cancer patients who underwent curative surgery. METHODS Fifty-eight patients underwent preoperative enhanced dynamic computed tomography (CT) scanning and fluorodeoxyglucose PET/CT using four PET scanners. Using LIFEx software, we measured PET radiomic parameters including texture features with higher order and harmonized these PET parameters. For progression-free survival (PFS) and overall survival (OS), we evaluated clinical information, including age, TNM stage, and neural invasion, and the harmonized PET radiomic features based on univariate Cox proportional hazard regression. Next, we analysed the prognostic indices by multivariate Cox proportional hazard regression (1) by using either significant (p < 0.05) or borderline significant (p = 0.05-0.10) indices in the univariate analysis (first multivariate analysis) or (2) by using the selected features with random forest algorithms (second multivariate analysis). Finally, we checked these multivariate results by log-rank test. RESULTS Regarding the first multivariate analysis for PFS after univariate analysis, age was the significant prognostic factor (p = 0.020), and MTV and GLCM contrast were borderline significant (p = 0.051 and 0.075, respectively). Regarding the first multivariate analysis of OS, neural invasion, Shape sphericity and GLZLM LZLGE were significant (p = 0.019, 0.042 and 0.0076). In the second multivariate analysis, only MTV was significant (p = 0.046) for PFS, whereas GLZLM LZLGE was significant (p = 0.047), and Shape sphericity was borderline significant (p = 0.088) for OS. In the log-rank test, age, MTV and GLCM contrast were borderline significant for PFS (p = 0.08, 0.06 and 0.07, respectively), whereas neural invasion and Shape sphericity were significant (p = 0.03 and 0.04, respectively), and GLZLM LZLGE was borderline significant for OS (p = 0.08). CONCLUSIONS Other than the clinical factors, MTV and GLCM contrast for PFS and Shape sphericity and GLZLM LZLGE for OS may be prognostic PET parameters. A prospective multicentre study with a larger sample size may be warranted.
Collapse
|
25
|
Spadarella G, Stanzione A, Akinci D'Antonoli T, Andreychenko A, Fanni SC, Ugga L, Kotter E, Cuocolo R. Systematic review of the radiomics quality score applications: an EuSoMII Radiomics Auditing Group Initiative. Eur Radiol 2023; 33:1884-1894. [PMID: 36282312 PMCID: PMC9935718 DOI: 10.1007/s00330-022-09187-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/31/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The main aim of the present systematic review was a comprehensive overview of the Radiomics Quality Score (RQS)-based systematic reviews to highlight common issues and challenges of radiomics research application and evaluate the relationship between RQS and review features. METHODS The literature search was performed on multiple medical literature archives according to PRISMA guidelines for systematic reviews that reported radiomic quality assessment through the RQS. Reported scores were converted to a 0-100% scale. The Mann-Whitney and Kruskal-Wallis tests were used to compare RQS scores and review features. RESULTS The literature research yielded 345 articles, from which 44 systematic reviews were finally included in the analysis. Overall, the median of RQS was 21.00% (IQR = 11.50). No significant differences of RQS were observed in subgroup analyses according to targets (oncological/not oncological target, neuroradiology/body imaging focus and one imaging technique/more than one imaging technique, characterization/prognosis/detection/other). CONCLUSIONS Our review did not reveal a significant difference of quality of radiomic articles reported in systematic reviews, divided in different subgroups. Furthermore, low overall methodological quality of radiomics research was found independent of specific application domains. While the RQS can serve as a reference tool to improve future study designs, future research should also be aimed at improving its reliability and developing new tools to meet an ever-evolving research space. KEY POINTS • Radiomics is a promising high-throughput method that may generate novel imaging biomarkers to improve clinical decision-making process, but it is an inherently complex analysis and often lacks reproducibility and generalizability. • The Radiomics Quality Score serves a necessary role as the de facto reference tool for assessing radiomics studies. • External auditing of radiomics studies, in addition to the standard peer-review process, is valuable to highlight common limitations and provide insights to improve future study designs and practical applicability of the radiomics models.
Collapse
Affiliation(s)
- Gaia Spadarella
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy.
| | - Tugba Akinci D'Antonoli
- Institute of Radiology and Nuclear Medicine, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Anna Andreychenko
- Research and Practical Clinical Center for Diagnostics and Telemedicine Technologies of the Moscow Healthcare Department, Moscow, Russia
| | | | - Lorenzo Ugga
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Elmar Kotter
- Department of Radiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Renato Cuocolo
- Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi, Italy
- Augmented Reality for Health Monitoring Laboratory (ARHeMLab), Department of Electrical Engineering and Information Technology, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
26
|
Predicting Soft Tissue Sarcoma Response to Neoadjuvant Chemotherapy Using an MRI-Based Delta-Radiomics Approach. Mol Imaging Biol 2023:10.1007/s11307-023-01803-y. [PMID: 36695966 DOI: 10.1007/s11307-023-01803-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023]
Abstract
OBJECTIVES To evaluate the performance of machine learning-augmented MRI-based radiomics models for predicting response to neoadjuvant chemotherapy (NAC) in soft tissue sarcomas. METHODS Forty-four subjects were identified retrospectively from patients who received NAC at our institution for pathologically proven soft tissue sarcomas. Only subjects who had both a baseline MRI prior to initiating chemotherapy and a post-treatment scan at least 2 months after initiating chemotherapy and prior to surgical resection were included. 3D ROIs were used to delineate whole-tumor volumes on pre- and post-treatment scans, from which 1708 radiomics features were extracted. Delta-radiomics features were calculated by subtraction of baseline from post-treatment values and used to distinguish treatment response through univariate analyses as well as machine learning-augmented radiomics analyses. RESULTS Though only 4.74% of variables overall reached significance at p ≤ 0.05 in univariate analyses, Laws Texture Energy (LTE)-derived metrics represented 46.04% of all such features reaching statistical significance. ROC analyses similarly failed to predict NAC response, with AUCs of 0.40 (95% CI 0.22-0.58) and 0.44 (95% CI 0.26-0.62) for RF and AdaBoost, respectively. CONCLUSION Overall, while our result was not able to separate NAC responders from non-responders, our analyses did identify a subset of LTE-derived metrics that show promise for further investigations. Future studies will likely benefit from larger sample size constructions so as to avoid the need for data filtering and feature selection techniques, which have the potential to significantly bias the machine learning procedures.
Collapse
|
27
|
Hannequin P, Decroisette C, Kermanach P, Berardi G, Bourbonne V. FDG PET and CT radiomics in diagnosis and prognosis of non-small-cell lung cancer. Transl Lung Cancer Res 2022; 11:2051-2063. [PMID: 36386457 PMCID: PMC9641045 DOI: 10.21037/tlcr-22-158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/22/2022] [Indexed: 09/13/2023]
Abstract
BACKGROUND 18F-FDG PET and CT radiomics has been the object of a wide research for over 20 years but its contribution to clinical practice remains not yet well established. We have investigated its impact versus that of only histo-clinical data, for the routine management of non-small-cell lung cancer (NSCLC). METHODS Our patients were retrospectively considered. They all had a FDG PET-CT and immuno-histo-chemistry (IHC) to assess PD-L1 expression at the beginning of the disease. A prognosis univariate and multivariate Cox survival analyses was performed for overall survival (OS) and progression free survival (PFS) prediction, including a training/testing procedure. Two sets of 47 PET and 47 CT radiomics features (RFs) were extracted. Difference between RFs according to PD-L1 expression, the histology status and the stage level were tested using suited non parametric statistical tests and the receiver operating characteristics (ROC) curve and the area under curve (AUC). RESULTS From 2017 to 2019, 212 NSCLC patients treated in our institution were included. The main conventional prognostic variables were stage and gender with a low added prognostic value in the models including PET and CT RFs. Neither PET nor CT RFs were significant to separate the different levels of PD-L1 expression. Several RFs differ between adenocarcinoma (ADC) and squamous cell carcinoma (SCC) tumours and a large number of PET and CT RFs are significantly linked to patient stage. CONCLUSIONS In our population, PET and CT RFs show their intrinsic power to predict survival but do not significantly improve OS and PFS prediction in the different multivariate models, in comparison to conventional data. It would seem necessary to carry out one's own survival analysis before determining a radiomics signature.
Collapse
Affiliation(s)
- Pascal Hannequin
- Annecy Nuclear Medicine Center, Le Pericles, B Allée de la Mandallaz, Metz-Tessy, France
| | - Chantal Decroisette
- Pneumology Department, CHANGE Annecy, 1 Avenue de l’hôpital, Metz-Tessy, France
| | - Pascale Kermanach
- Mont Blanc Histo-Pathology Laboratory, 40 Route de l’Aiglière, Argonay, France
| | - Giulia Berardi
- Pneumology Department, University Hospital la Tronche, Boulevard de la Chantourne, La Tronche, France
| | - Vincent Bourbonne
- Radiation Oncology Department, University Hospital, 2 Avenue Foch, Brest, France
| |
Collapse
|
28
|
Noortman WA, Vriens D, de Geus-Oei LF, Slump CH, Aarntzen EH, van Berkel A, Timmers HJLM, van Velden FHP. [ 18F]FDG-PET/CT radiomics for the identification of genetic clusters in pheochromocytomas and paragangliomas. Eur Radiol 2022; 32:7227-7236. [PMID: 36001126 PMCID: PMC9474528 DOI: 10.1007/s00330-022-09034-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/15/2022] [Accepted: 07/16/2022] [Indexed: 11/04/2022]
Abstract
Objectives Based on germline and somatic mutation profiles, pheochromocytomas and paragangliomas (PPGLs) can be classified into different clusters. We investigated the use of [18F]FDG-PET/CT radiomics, SUVmax and biochemical profile for the identification of the genetic clusters of PPGLs. Methods In this single-centre cohort, 40 PPGLs (13 cluster 1, 18 cluster 2, 9 sporadic) were delineated using a 41% adaptive threshold of SUVpeak ([18F]FDG-PET) and manually (low-dose CT; ldCT). Using PyRadiomics, 211 radiomic features were extracted. Stratified 5-fold cross-validation for the identification of the genetic cluster was performed using multinomial logistic regression with dimensionality reduction incorporated per fold. Classification performances of biochemistry, SUVmax and PET(/CT) radiomic models were compared and presented as mean (multiclass) test AUCs over the five folds. Results were validated using a sham experiment, randomly shuffling the outcome labels. Results The model with biochemistry only could identify the genetic cluster (multiclass AUC 0.60). The three-factor PET model had the best classification performance (multiclass AUC 0.88). A simplified model with only SUVmax performed almost similarly. Addition of ldCT features and biochemistry decreased the classification performances. All sham AUCs were approximately 0.50. Conclusion PET radiomics achieves a better identification of PPGLs compared to biochemistry, SUVmax, ldCT radiomics and combined approaches, especially for the differentiation of sporadic PPGLs. Nevertheless, a model with SUVmax alone might be preferred clinically, weighing model performances against laborious radiomic analysis. The limited added value of radiomics to the overall classification performance for PPGL should be validated in a larger external cohort. Key Points • Radiomics derived from [18F]FDG-PET/CT has the potential to improve the identification of the genetic clusters of pheochromocytomas and paragangliomas. • A simplified model with SUVmaxonly might be preferred clinically, weighing model performances against the laborious radiomic analysis. • Cluster 1 and 2 PPGLs generally present distinctive characteristics that can be captured using [18F]FDG-PET imaging. Sporadic PPGLs appear more heterogeneous, frequently resembling cluster 2 PPGLs and occasionally resembling cluster 1 PPGLs. Supplementary Information The online version contains supplementary material available at 10.1007/s00330-022-09034-5.
Collapse
Affiliation(s)
- Wyanne A Noortman
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands. .,TechMed Centre, University of Twente, Enschede, the Netherlands.
| | - Dennis Vriens
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands.,TechMed Centre, University of Twente, Enschede, the Netherlands.,Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Erik H Aarntzen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anouk van Berkel
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Henri J L M Timmers
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Floris H P van Velden
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
29
|
Stanzione A, Galatola R, Cuocolo R, Romeo V, Verde F, Mainenti PP, Brunetti A, Maurea S. Radiomics in Cross-Sectional Adrenal Imaging: A Systematic Review and Quality Assessment Study. Diagnostics (Basel) 2022; 12:diagnostics12030578. [PMID: 35328133 PMCID: PMC8947112 DOI: 10.3390/diagnostics12030578] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 12/22/2022] Open
Abstract
In this study, we aimed to systematically review the current literature on radiomics applied to cross-sectional adrenal imaging and assess its methodological quality. Scopus, PubMed and Web of Science were searched to identify original research articles investigating radiomics applications on cross-sectional adrenal imaging (search end date February 2021). For qualitative synthesis, details regarding study design, aim, sample size and imaging modality were recorded as well as those regarding the radiomics pipeline (e.g., segmentation and feature extraction strategy). The methodological quality of each study was evaluated using the radiomics quality score (RQS). After duplicate removal and selection criteria application, 25 full-text articles were included and evaluated. All were retrospective studies, mostly based on CT images (17/25, 68%), with manual (19/25, 76%) and two-dimensional segmentation (13/25, 52%) being preferred. Machine learning was paired to radiomics in about half of the studies (12/25, 48%). The median total and percentage RQS scores were 2 (interquartile range, IQR = −5–8) and 6% (IQR = 0–22%), respectively. The highest and lowest scores registered were 12/36 (33%) and −5/36 (0%). The most critical issues were the absence of proper feature selection, the lack of appropriate model validation and poor data openness. The methodological quality of radiomics studies on adrenal cross-sectional imaging is heterogeneous and lower than desirable. Efforts toward building higher quality evidence are essential to facilitate the future translation into clinical practice.
Collapse
Affiliation(s)
- Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (A.S.); (R.G.); (V.R.); (F.V.); (A.B.); (S.M.)
| | - Roberta Galatola
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (A.S.); (R.G.); (V.R.); (F.V.); (A.B.); (S.M.)
| | - Renato Cuocolo
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy
- Interdepartmental Research Center on Management and Innovation in Healthcare-CIRMIS, University of Naples “Federico II”, 80100 Naples, Italy
- Laboratory of Augmented Reality for Health Monitoring (ARHeMLab), Department of Electrical Engineering and Information Technology, University of Naples “Federico II”, 80100 Naples, Italy
- Correspondence:
| | - Valeria Romeo
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (A.S.); (R.G.); (V.R.); (F.V.); (A.B.); (S.M.)
| | - Francesco Verde
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (A.S.); (R.G.); (V.R.); (F.V.); (A.B.); (S.M.)
| | - Pier Paolo Mainenti
- Institute of Biostructures and Bioimaging of the National Research Council, 80131 Naples, Italy;
| | - Arturo Brunetti
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (A.S.); (R.G.); (V.R.); (F.V.); (A.B.); (S.M.)
| | - Simone Maurea
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (A.S.); (R.G.); (V.R.); (F.V.); (A.B.); (S.M.)
| |
Collapse
|
30
|
Anan N, Zainon R, Tamal M. A review on advances in 18F-FDG PET/CT radiomics standardisation and application in lung disease management. Insights Imaging 2022; 13:22. [PMID: 35124733 PMCID: PMC8817778 DOI: 10.1186/s13244-021-01153-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Radiomics analysis quantifies the interpolation of multiple and invisible molecular features present in diagnostic and therapeutic images. Implementation of 18-fluorine-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) radiomics captures various disorders in non-invasive and high-throughput manner. 18F-FDG PET/CT accurately identifies the metabolic and anatomical changes during cancer progression. Therefore, the application of 18F-FDG PET/CT in the field of oncology is well established. Clinical application of 18F-FDG PET/CT radiomics in lung infection and inflammation is also an emerging field. Combination of bioinformatics approaches or textual analysis allows radiomics to extract additional information to predict cell biology at the micro-level. However, radiomics texture analysis is affected by several factors associated with image acquisition and processing. At present, researchers are working on mitigating these interrupters and developing standardised workflow for texture biomarker establishment. This review article focuses on the application of 18F-FDG PET/CT in detecting lung diseases specifically on cancer, infection and inflammation. An overview of different approaches and challenges encountered on standardisation of 18F-FDG PET/CT technique has also been highlighted. The review article provides insights about radiomics standardisation and application of 18F-FDG PET/CT in lung disease management.
Collapse
|
31
|
Kolinger GD, Vállez García D, Kramer GM, Frings V, Zwezerijnen GJC, Smit EF, De Langen AJ, Buvat I, Boellaard R. Effects of tracer uptake time in non-small cell lung cancer 18F-FDG PET radiomics. J Nucl Med 2021; 63:919-924. [PMID: 34933890 DOI: 10.2967/jnumed.121.262660] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/21/2021] [Indexed: 11/16/2022] Open
Abstract
Positron emission tomography (PET) radiomics applied to oncology allows the measurement of intra-tumoral heterogeneity. This quantification can be affected by image protocols hence there is an increased interest in understanding how radiomic expression on PET images is affected by different imaging conditions. To address that, this study explores how radiomic features are affected by changes in 18F-FDG uptake time, image reconstruction, lesion delineation, and radiomics binning settings. Methods: Ten non-small cell lung cancer (NSCLC) patients underwent 18F-FDG PET scans on two consecutive days. On each day, scans were obtained at 60min and 90min post-injection and reconstructed following EARL version 1 (EARL1) and with point-spread-function resolution modelling (PSF-EARL2). Lesions were delineated using thresholds at SUV=4.0, 40% of SUVmax, and with a contrast-based isocontour. PET image intensity was discretized with both fixed bin width (FBW) and fixed bin number (FBN) before the calculation of the radiomic features. Repeatability of features was measured with intraclass correlation (ICC), and the change in feature value over time was calculated as a function of its repeatability. Features were then classified on use-case scenarios based on their repeatability and susceptibility to tracer uptake time. Results: With PSF-EARL2 reconstruction, 40% of SUVmax lesion delineation, and FBW intensity discretization, most features (94%) were repeatable at both uptake times (ICC>0.9), 39% being classified for dual-time-point use-case for being sensitive to changes in uptake time, 39% were classified for cross-sectional studies with unclear dependency on time, 20% classified for cross-sectional use while being robust to tracer uptake time changes, and 6% were discarded for poor repeatability. EARL1 images had one less repeatable feature than PSF-EARL2 (Neighborhood Gray-Level Different Matrix Coarseness), the contrast-based delineation had the poorest repeatability of the delineation methods with 45% features being discarded, and FBN resulted in lower repeatability than FBW (45% and 6% features were discarded, respectively). Conclusion: Repeatability was maximized with PSF-EARL2 reconstruction, lesion delineation at 40% of SUVmax, and FBW intensity discretization. Based on their susceptibility to tracer uptake time, radiomic features were classified into specific NSCLC PET radiomics use-cases.
Collapse
Affiliation(s)
| | - David Vállez García
- Medical Imaging Center, University Medical Center Groningen, University of Groningen, Netherlands
| | - Gerbrand Maria Kramer
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, location VU Medical Center, Netherlands
| | - Virginie Frings
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, location VU Medical Center, Netherlands
| | | | - Egbert F Smit
- Department of Pulmonology, Amsterdam University Medical Center, location VU Medical Center, Netherlands
| | | | - Irène Buvat
- Laboratoire d'Imagerie Translationnelle en Oncologie, INSERM, Institut Curie, Université Paris-Saclay, France
| | - Ronald Boellaard
- Medical Imaging Center, University Medical Center Groningen, University of Groningen, Netherlands
| |
Collapse
|
32
|
Taghavi M, Staal FCR, Simões R, Hong EK, Lambregts DMJ, van der Heide UA, Beets-Tan RGH, Maas M. CT radiomics models are unable to predict new liver metastasis after successful thermal ablation of colorectal liver metastases. Acta Radiol 2021; 64:5-12. [PMID: 34918955 DOI: 10.1177/02841851211060437] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Patients with colorectal liver metastases (CRLM) who undergo thermal ablation are at risk of developing new CRLM after ablation. Identification of these patients might enable individualized treatment. PURPOSE To investigate whether an existing machine-learning model with radiomics features based on pre-ablation computed tomography (CT) images of patients with colorectal cancer can predict development of new CRLM. MATERIAL AND METHODS In total, 94 patients with CRLM who were treated with thermal ablation were analyzed. Radiomics features were extracted from the healthy liver parenchyma of CT images in the portal venous phase, before thermal ablation. First, a previously developed radiomics model (Original model) was applied to the entire cohort to predict new CRLM after 6 and 24 months of follow-up. Next, new machine-learning models were developed (Radiomics, Clinical, and Combined), based on radiomics features, clinical features, or a combination of both. RESULTS The external validation of the Original model reached an area under the curve (AUC) of 0.57 (95% confidence interval [CI]=0.56-0.58) and 0.52 (95% CI=0.51-0.53) for 6 and 24 months of follow-up. The new predictive radiomics models yielded a higher performance at 6 months compared to 24 months. For the prediction of CRLM at 6 months, the Combined model had slightly better performance (AUC=0.60; 95% CI=0.59-0.61) compared to the Radiomics and Clinical models (AUC=0.55-0.57), while all three models had a low performance for the prediction at 24 months (AUC=0.52-0.53). CONCLUSION Both the Original and newly developed radiomics models were unable to predict new CLRM based on healthy liver parenchyma in patients who will undergo ablation for CRLM.
Collapse
Affiliation(s)
- Marjaneh Taghavi
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School of Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Femke CR Staal
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School of Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Rita Simões
- Department of Radiotherapy, Netherland Cancer Institute, Amsterdam, The Netherlands
| | - Eun K Hong
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School of Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
- Seoul National University Hospital, Seoul, Republic of Korea
| | - Doenja MJ Lambregts
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Regina GH Beets-Tan
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School of Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
- Institute of Regional Health Research, University of Southern Denmark, Denmark
| | - Monique Maas
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Ahrari S, Zaragori T, Rozenblum L, Oster J, Imbert L, Kas A, Verger A. Relevance of Dynamic 18F-DOPA PET Radiomics for Differentiation of High-Grade Glioma Progression from Treatment-Related Changes. Biomedicines 2021; 9:biomedicines9121924. [PMID: 34944740 PMCID: PMC8698938 DOI: 10.3390/biomedicines9121924] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 12/22/2022] Open
Abstract
This study evaluates the relevance of 18F-DOPA PET static and dynamic radiomics for differentiation of high-grade glioma (HGG) progression from treatment-related changes (TRC) by comparing diagnostic performances to the current PET imaging standard of care. Eighty-five patients with histologically confirmed HGG and investigated by dynamic 18F-FDOPA PET in two institutions were retrospectively selected. ElasticNet logistic regression, Random Forest and XGBoost machine models were trained with different sets of features-radiomics extracted from static tumor-to-background-ratio (TBR) parametric images, radiomics extracted from time-to-peak (TTP) parametric images, as well as combination of both-in order to discriminate glioma progression from TRC at 6 months from the PET scan. Diagnostic performances of the models were compared to a logistic regression model with TBRmean ± clinical features used as reference. Training was performed on data from the first center, while external validation was performed on data from the second center. Best radiomics models showed only slightly better performances than the reference model (respective AUCs of 0.834 vs. 0.792, p < 0.001). Our current results show similar findings at the multicentric level using different machine learning models and report a marginal additional value for TBR static and TTP dynamic radiomics over the classical analysis based on TBR values.
Collapse
Affiliation(s)
- Shamimeh Ahrari
- Université de Lorraine, IADI, INSERM, UMR 1254, F-54000 Nancy, France; (S.A.); (T.Z.); (J.O.); (L.I.)
| | - Timothée Zaragori
- Université de Lorraine, IADI, INSERM, UMR 1254, F-54000 Nancy, France; (S.A.); (T.Z.); (J.O.); (L.I.)
| | - Laura Rozenblum
- Sorbonne Université, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Service de Médecine Nucléaire and LIB, INSERM U1146, F-75013 Paris, France; (L.R.); (A.K.)
| | - Julien Oster
- Université de Lorraine, IADI, INSERM, UMR 1254, F-54000 Nancy, France; (S.A.); (T.Z.); (J.O.); (L.I.)
| | - Laëtitia Imbert
- Université de Lorraine, IADI, INSERM, UMR 1254, F-54000 Nancy, France; (S.A.); (T.Z.); (J.O.); (L.I.)
- Department of Nuclear Medicine & Nancyclotep Imaging Platform, Université de Lorraine, CHRU-Nancy, F-54000 Nancy, France
| | - Aurélie Kas
- Sorbonne Université, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Service de Médecine Nucléaire and LIB, INSERM U1146, F-75013 Paris, France; (L.R.); (A.K.)
| | - Antoine Verger
- Université de Lorraine, IADI, INSERM, UMR 1254, F-54000 Nancy, France; (S.A.); (T.Z.); (J.O.); (L.I.)
- Department of Nuclear Medicine & Nancyclotep Imaging Platform, Université de Lorraine, CHRU-Nancy, F-54000 Nancy, France
- Correspondence:
| |
Collapse
|
34
|
Edalat-Javid M, Shiri I, Hajianfar G, Abdollahi H, Arabi H, Oveisi N, Javadian M, Shamsaei Zafarghandi M, Malek H, Bitarafan-Rajabi A, Oveisi M, Zaidi H. Cardiac SPECT radiomic features repeatability and reproducibility: A multi-scanner phantom study. J Nucl Cardiol 2021; 28:2730-2744. [PMID: 32333282 DOI: 10.1007/s12350-020-02109-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/12/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND The aim of this work was to assess the robustness of cardiac SPECT radiomic features against changes in imaging settings, including acquisition, and reconstruction parameters. METHODS Four commercial SPECT and SPECT/CT cameras were used to acquire images of a static cardiac phantom mimicking typical myorcardial perfusion imaging using 185 MBq of 99mTc. The effects of different image acquisition and reconstruction parameters, including number of views, view matrix size, attenuation correction, as well as image reconstruction related parameters (algorithm, number of iterations, number of subsets, type of post-reconstruction filter, and its associated parameters, including filter order and cut-off frequency) were studied. In total, 5,063 transverse views were reconstructed by varying the aforementioned factors. Eighty-seven radiomic features including first-, second-, and high-order textures were extracted from these images. To assess reproducibility and repeatability, the coefficient of variation (COV), as a widely adopted metric, was measured for each of the radiomic features over the different imaging settings. RESULTS The Inverse Difference Moment Normalized (IDMN) and Inverse Difference Normalized (IDN) features from the Gray Level Co-occurrence Matrix (GLCM), Run Percentage (RP) from the Gray Level Co-occurrence Matrix (GLRLM), Zone Entropy (ZE) from the Gray Level Size Zone Matrix (GLSZM), and Dependence Entropy (DE) from the Gray Level Dependence Matrix (GLDM) feature sets were the only features that exhibited high reproducibility (COV ≤ 5%) against changes in all imaging settings. In addition, Large Area Low Gray Level Emphasis (LALGLE), Small Area Low Gray Level Emphasis (SALGLE) and Low Gray Level Zone Emphasis (LGLZE) from GLSZM, and Small Dependence Low Gray Level Emphasis (SDLGLE) from GLDM feature sets turned out to be less reproducible (COV > 20%) against changes in imaging settings. The GLRLM (31.88%) and GLDM feature set (54.2%) had the highest (COV < 5%) and lowest (COV > 20%) number of the reproducible features, respectively. Matrix size had the largest impact on feature variability as most of the features were not repeatable when matrix size was modified with 82.8% of them having a COV > 20%. CONCLUSION The repeatability and reproducibility of SPECT/CT cardiac radiomic features under different imaging settings is feature-dependent. Different image acquisition and reconstruction protocols have variable effects on radiomic features. The radiomic features exhibiting low COV are potential candidates for future clinical studies.
Collapse
Affiliation(s)
- Mohammad Edalat-Javid
- Department of Energy Engineering and Physics, Amir Kabir University of Technology, Tehran, Iran
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, 1211, Geneva 4, Switzerland
| | - Ghasem Hajianfar
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| | - Hamid Abdollahi
- Department of Radiologic Sciences and Medical Physics, Faculty of Allied Medicine, Kerman University, Kerman, Iran
| | - Hossein Arabi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, 1211, Geneva 4, Switzerland
| | - Niki Oveisi
- School of Population and Public Health, The University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Mohammad Javadian
- Department of Computer Engineering, Faculty of Information Technology, Kermanshah University of Technology, Kermanshah, Iran
| | | | - Hadi Malek
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| | - Ahmad Bitarafan-Rajabi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
- Cardiovascular Intervention Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
- Echocardiography Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Oveisi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
- Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, 1211, Geneva 4, Switzerland.
- Geneva University Neurocenter, Geneva University, 1205, 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.
| |
Collapse
|
35
|
Pfaehler E, Zhovannik I, Wei L, Boellaard R, Dekker A, Monshouwer R, El Naqa I, Bussink J, Gillies R, Wee L, Traverso A. A systematic review and quality of reporting checklist for repeatability and reproducibility of radiomic features. Phys Imaging Radiat Oncol 2021; 20:69-75. [PMID: 34816024 PMCID: PMC8591412 DOI: 10.1016/j.phro.2021.10.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022] Open
Abstract
Main factors impacting feature stability: Image acquisition, reconstruction, tumor segmentation, and interpolation. Textural features are less robust than morphological or statistical features. A checklist is provided including items that should be reported in a radiomic study.
Purpose Although quantitative image biomarkers (radiomics) show promising value for cancer diagnosis, prognosis, and treatment assessment, these biomarkers still lack reproducibility. In this systematic review, we aimed to assess the progress in radiomics reproducibility and repeatability in the recent years. Methods and materials Four hundred fifty-one abstracts were retrieved according to the original PubMed search pattern with the publication dates ranging from 2017/05/01 to 2020/12/01. Each abstract including the keywords was independently screened by four observers. Forty-two full-text articles were selected for further analysis. Patient population data, radiomic feature classes, feature extraction software, image preprocessing, and reproducibility results were extracted from each article. To support the community with a standardized reporting strategy, we propose a specific reporting checklist to evaluate the feasibility to reproduce each study. Results Many studies continue to under-report essential reproducibility information: all but one clinical and all but two phantom studies missed to report at least one important item reporting image acquisition. The studies included in this review indicate that all radiomic features are sensitive to image acquisition, reconstruction, tumor segmentation, and interpolation. However, the amount of sensitivity is feature dependent, for instance, textural features were, in general, less robust than statistical features. Conclusions Radiomics repeatability, reproducibility, and reporting quality can substantially be improved regarding feature extraction software and settings, image preprocessing and acquisition, cutoff values for stable feature selection. Our proposed radiomics reporting checklist can serve to simplify and improve the reporting and, eventually, guarantee the possibility to fully replicate and validate radiomic studies.
Collapse
Affiliation(s)
- Elisabeth Pfaehler
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ivan Zhovannik
- Department of Radiation Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Radiation Oncology (MAASTRO), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Lise Wei
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - René Monshouwer
- Department of Radiation Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Issam El Naqa
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Jan Bussink
- Department of Radiation Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Robert Gillies
- Department of Radiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Leonard Wee
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Alberto Traverso
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
36
|
Orlhac F, Nioche C, Klyuzhin I, Rahmim A, Buvat I. Radiomics in PET Imaging:: A Practical Guide for Newcomers. PET Clin 2021; 16:597-612. [PMID: 34537132 DOI: 10.1016/j.cpet.2021.06.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Radiomics has undergone considerable development in recent years. In PET imaging, very promising results concerning the ability of handcrafted features to predict the biological characteristics of lesions and to assess patient prognosis or response to treatment have been reported in the literature. This article presents a checklist for designing a reliable radiomic study, gives an overview of the steps of the pipeline, and outlines approaches for data harmonization. Tips are provided for critical reading of the content of articles. The advantages and limitations of handcrafted radiomics compared with deep-learning approaches for the characterization of PET images are also discussed.
Collapse
Affiliation(s)
- Fanny Orlhac
- Institut Curie Centre de Recherche, Centre Universitaire, Bat 101B, Rue Henri Becquerel, CS 90030, 91401 Orsay Cedex, France.
| | - Christophe Nioche
- Institut Curie Centre de Recherche, Centre Universitaire, Bat 101B, Rue Henri Becquerel, CS 90030, 91401 Orsay Cedex, France
| | - Ivan Klyuzhin
- Department of Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada; Department of Radiology, University of British Columbia, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada; Department of Radiology, University of British Columbia, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada
| | - Irène Buvat
- Institut Curie Centre de Recherche, Centre Universitaire, Bat 101B, Rue Henri Becquerel, CS 90030, 91401 Orsay Cedex, France
| |
Collapse
|
37
|
Stanzione A, Verde F, Romeo V, Boccadifuoco F, Mainenti PP, Maurea S. Radiomics and machine learning applications in rectal cancer: Current update and future perspectives. World J Gastroenterol 2021; 27:5306-5321. [PMID: 34539134 PMCID: PMC8409167 DOI: 10.3748/wjg.v27.i32.5306] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/13/2021] [Accepted: 07/22/2021] [Indexed: 02/06/2023] Open
Abstract
The high incidence of rectal cancer in both sexes makes it one of the most common tumors, with significant morbidity and mortality rates. To define the best treatment option and optimize patient outcome, several rectal cancer biological variables must be evaluated. Currently, medical imaging plays a crucial role in the characterization of this disease, and it often requires a multimodal approach. Magnetic resonance imaging is the first-choice imaging modality for local staging and restaging and can be used to detect high-risk prognostic factors. Computed tomography is widely adopted for the detection of distant metastases. However, conventional imaging has recognized limitations, and many rectal cancer characteristics remain assessable only after surgery and histopathology evaluation. There is a growing interest in artificial intelligence applications in medicine, and imaging is by no means an exception. The introduction of radiomics, which allows the extraction of quantitative features that reflect tumor heterogeneity, allows the mining of data in medical images and paved the way for the identification of potential new imaging biomarkers. To manage such a huge amount of data, the use of machine learning algorithms has been proposed. Indeed, without prior explicit programming, they can be employed to build prediction models to support clinical decision making. In this review, current applications and future perspectives of artificial intelligence in medical imaging of rectal cancer are presented, with an imaging modality-based approach and a keen eye on unsolved issues. The results are promising, but the road ahead for translation in clinical practice is rather long.
Collapse
Affiliation(s)
- Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples 80131, Italy
| | - Francesco Verde
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples 80131, Italy
| | - Valeria Romeo
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples 80131, Italy
| | - Francesca Boccadifuoco
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples 80131, Italy
| | - Pier Paolo Mainenti
- Institute of Biostructures and Bioimaging, National Council of Research, Napoli 80131, Italy
| | - Simone Maurea
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples 80131, Italy
| |
Collapse
|
38
|
The Influence of the Exclusion of Central Necrosis on [ 18F]FDG PET Radiomic Analysis. Diagnostics (Basel) 2021; 11:diagnostics11071296. [PMID: 34359379 PMCID: PMC8304274 DOI: 10.3390/diagnostics11071296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Central necrosis can be detected on [18F]FDG PET/CT as a region with little to no tracer uptake. Currently, there is no consensus regarding the inclusion of regions of central necrosis during volume of interest (VOI) delineation for radiomic analysis. The aim of this study was to assess how central necrosis affects radiomic analysis in PET. Methods: Forty-three patients, either with non-small cell lung carcinomas (NSCLC, n = 12) or with pheochromocytomas or paragangliomas (PPGL, n = 31), were included retrospectively. VOIs were delineated with and without central necrosis. From all VOIs, 105 radiomic features were extracted. Differences in radiomic features between delineation methods were assessed using a paired t-test with Benjamini–Hochberg multiple testing correction. In the PPGL cohort, performances of the radiomic models to predict the noradrenergic biochemical profile were assessed by comparing the areas under the receiver operating characteristic curve (AUC) for both delineation methods. Results: At least 65% of the features showed significant differences between VOIvital-tumour and VOIgross-tumour (65%, 79% and 82% for the NSCLC, PPGL and combined cohort, respectively). The AUCs of the radiomic models were not significantly different between delineation methods. Conclusion: In both tumour types, almost two-third of the features were affected, demonstrating that the impact of whether or not to include central necrosis in the VOI on the radiomic feature values is significant. Nevertheless, predictive performances of both delineation methods were comparable. We recommend that radiomic studies should report whether or not central necrosis was included during delineation.
Collapse
|
39
|
Noortman WA, Vriens D, Mooij CDY, Slump CH, Aarntzen EH, van Berkel A, Timmers HJLM, Bussink J, Meijer TWH, de Geus-Oei LF, van Velden FHP. The Influence of the Exclusion of Central Necrosis on [ 18F]FDG PET Radiomic Analysis. DIAGNOSTICS (BASEL, SWITZERLAND) 2021. [PMID: 34359379 DOI: 10.3390/diagnostics] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Central necrosis can be detected on [18F]FDG PET/CT as a region with little to no tracer uptake. Currently, there is no consensus regarding the inclusion of regions of central necrosis during volume of interest (VOI) delineation for radiomic analysis. The aim of this study was to assess how central necrosis affects radiomic analysis in PET. METHODS Forty-three patients, either with non-small cell lung carcinomas (NSCLC, n = 12) or with pheochromocytomas or paragangliomas (PPGL, n = 31), were included retrospectively. VOIs were delineated with and without central necrosis. From all VOIs, 105 radiomic features were extracted. Differences in radiomic features between delineation methods were assessed using a paired t-test with Benjamini-Hochberg multiple testing correction. In the PPGL cohort, performances of the radiomic models to predict the noradrenergic biochemical profile were assessed by comparing the areas under the receiver operating characteristic curve (AUC) for both delineation methods. RESULTS At least 65% of the features showed significant differences between VOIvital-tumour and VOIgross-tumour (65%, 79% and 82% for the NSCLC, PPGL and combined cohort, respectively). The AUCs of the radiomic models were not significantly different between delineation methods. CONCLUSION In both tumour types, almost two-third of the features were affected, demonstrating that the impact of whether or not to include central necrosis in the VOI on the radiomic feature values is significant. Nevertheless, predictive performances of both delineation methods were comparable. We recommend that radiomic studies should report whether or not central necrosis was included during delineation.
Collapse
Affiliation(s)
- Wyanne A Noortman
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Dennis Vriens
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Charlotte D Y Mooij
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Technical Medicine, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Cornelis H Slump
- TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Erik H Aarntzen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Anouk van Berkel
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Henri J L M Timmers
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Tineke W H Meijer
- Department of Radiation Oncology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Floris H P van Velden
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
40
|
Cucchiara F, Petrini I, Romei C, Crucitta S, Lucchesi M, Valleggi S, Scavone C, Capuano A, De Liperi A, Chella A, Danesi R, Del Re M. Combining liquid biopsy and radiomics for personalized treatment of lung cancer patients. State of the art and new perspectives. Pharmacol Res 2021; 169:105643. [PMID: 33940185 DOI: 10.1016/j.phrs.2021.105643] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022]
Abstract
Lung cancer has become a paradigm for precision medicine in oncology, and liquid biopsy (LB) together with radiomics may have a great potential in this scenario. They are both minimally invasive, easy to perform, and can be repeated during patient's follow-up. Also, increasing evidence suggest that LB and radiomics may provide an efficient way to screen and diagnose tumors at an early stage, including the monitoring of any change in the tumor molecular profile. This could allow treatment optimization, improvement of patients' quality of life, and healthcare-related costs reduction. Latest reports on lung cancer patients suggest a combination of these two strategies, along with cutting-edge data analysis, to decode valuable information regarding tumor type, aggressiveness, progression, and response to treatment. The approach seems more compatible with clinical practice than the current standard, and provides new diagnostic companions being able to suggest the best treatment strategy compared to conventional methods. To implement radiomics and liquid biopsy directly into clinical practice, an artificial intelligence (AI)-based system could help to link patients' clinical data together with tumor molecular profiles and imaging characteristics. AI could also solve problems and limitations related to LB and radiomics methodologies. Further work is needed, including new health policies and the access to large amounts of high-quality and well-organized data, allowing a complementary and synergistic combination of LB and imaging, to provide an attractive choice e in the personalized treatment of lung cancer.
Collapse
Affiliation(s)
- Federico Cucchiara
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Iacopo Petrini
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Chiara Romei
- Unit II of Radio-diagnostics, Department of Diagnostic and Imaging, University Hospital of Pisa, Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Maurizio Lucchesi
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Simona Valleggi
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Cristina Scavone
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annalisa De Liperi
- Unit II of Radio-diagnostics, Department of Diagnostic and Imaging, University Hospital of Pisa, Pisa, Italy
| | - Antonio Chella
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy.
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
41
|
Angus L, Starmans MPA, Rajicic A, Odink AE, Jalving M, Niessen WJ, Visser JJ, Sleijfer S, Klein S, van der Veldt AAM. The BRAF P.V600E Mutation Status of Melanoma Lung Metastases Cannot Be Discriminated on Computed Tomography by LIDC Criteria nor Radiomics Using Machine Learning. J Pers Med 2021; 11:257. [PMID: 33915880 PMCID: PMC8066683 DOI: 10.3390/jpm11040257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 11/05/2022] Open
Abstract
Patients with BRAF mutated (BRAF-mt) metastatic melanoma benefit significantly from treatment with BRAF inhibitors. Currently, the BRAF status is determined on archival tumor tissue or on fresh tumor tissue from an invasive biopsy. The aim of this study was to evaluate whether radiomics can predict the BRAF status in a non-invasive manner. Patients with melanoma lung metastases, known BRAF status, and a pretreatment computed tomography scan were included. After semi-automatic annotation of the lung lesions (maximum two per patient), 540 radiomics features were extracted. A chest radiologist scored all segmented lung lesions according to the Lung Image Database Consortium (LIDC) criteria. Univariate analysis was performed to assess the predictive value of each feature for BRAF mutation status. A combination of various machine learning methods was used to develop BRAF decision models based on the radiomics features and LIDC criteria. A total of 169 lung lesions from 103 patients (51 BRAF-mt; 52 BRAF wild type) were included. There were no features with a significant discriminative value in the univariate analysis. Models based on radiomics features and LIDC criteria both performed as poorly as guessing. Hence, the BRAF mutation status in melanoma lung metastases cannot be predicted using radiomics features or visually scored LIDC criteria.
Collapse
Affiliation(s)
- Lindsay Angus
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (A.R.); (S.S.); (A.A.M.v.d.V.)
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (M.P.A.S.); (A.E.O.); (W.J.N.); (J.J.V.); (S.K.)
| | - Martijn P. A. Starmans
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (M.P.A.S.); (A.E.O.); (W.J.N.); (J.J.V.); (S.K.)
- Department of Medical Informatics, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Ana Rajicic
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (A.R.); (S.S.); (A.A.M.v.d.V.)
| | - Arlette E. Odink
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (M.P.A.S.); (A.E.O.); (W.J.N.); (J.J.V.); (S.K.)
| | - Mathilde Jalving
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| | - Wiro J. Niessen
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (M.P.A.S.); (A.E.O.); (W.J.N.); (J.J.V.); (S.K.)
- Department of Medical Informatics, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Faculty of Applied Sciences, Delft University of Technology, 2628 CJ Delft, The Netherlands
| | - Jacob J. Visser
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (M.P.A.S.); (A.E.O.); (W.J.N.); (J.J.V.); (S.K.)
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (A.R.); (S.S.); (A.A.M.v.d.V.)
| | - Stefan Klein
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (M.P.A.S.); (A.E.O.); (W.J.N.); (J.J.V.); (S.K.)
- Department of Medical Informatics, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Astrid A. M. van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (A.R.); (S.S.); (A.A.M.v.d.V.)
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (M.P.A.S.); (A.E.O.); (W.J.N.); (J.J.V.); (S.K.)
| |
Collapse
|
42
|
Adhesion of Electrospun Poly(acrylonitrile) Nanofibers on Conductive and Isolating Foil Substrates. COATINGS 2021. [DOI: 10.3390/coatings11020249] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Electrospinning can be used to prepare nanofibers from various polymers and polymer blends. The adhesion of nanofibers to the substrates on which they are electrospun varies greatly with the substrate material and structure. In some cases, good adhesion is desired to produce sandwich structures by electrospinning one material directly onto another. This is the case, e.g., with dye-sensitized solar cells (DSSCs). While both pure foil DSSCs and pure electrospun DSSCs have been examined, a combination of both technologies can be used to combine their advantages, e.g., the lateral strength of foils with the large surface-to-volume ratio of electrospun nanofibers. Here, we investigate the morphology and adhesion of electrospun nanofibers on different foil substrates containing materials commonly used in DSSCs, such as graphite, poly(3,4-ethylenedioxythiophene) polystyrene sulfonate (PEDOT:PSS) or TiO2. The results show that the foil material strongly influences the adhesion, while a plasma pretreatment of the foils showed no significant effect. Moreover, it is well known that conductive substrates can alter the morphology of nanofiber mats, both at microscopic and macroscopic levels. However, these effects could not be observed in the current study.
Collapse
|
43
|
Crombé A, Buy X, Han F, Toupin S, Kind M. Assessment of Repeatability, Reproducibility, and Performances of T2 Mapping-Based Radiomics Features: A Comparative Study. J Magn Reson Imaging 2021; 54:537-548. [PMID: 33594768 DOI: 10.1002/jmri.27558] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Magnetic resonance imaging (MRI)-based radiomics features (RFs) quantify tumors radiological phenotypes but are sensitive to postprocessing parameters, including the intensity harmonization technique (IHT), while mappings enable objective quantitative assessment. PURPOSE To investigate whether T2 mapping could improve repeatability, reproducibility, and performances of radiomics compared to conventional T2-weighted imaging (T2WI). STUDY TYPE Prospective. SUBJECTS Twenty-six healthy adults. FIELD STRENGTH/SEQUENCE Respiratory-trigged radial turbo spin echo (TSE) multiecho T2 mapping (prototype) and conventional TSE T2WI of the abdomen were acquired twice at 1.5 T. ASSESSMENT T2 maps were reconstructed using a two-parameter exponential fitting model. Volumes-of-interest (VOIs) were manually drawn in six tissues: liver, kidney, pancreas, muscle, bone, and spleen. After co-registration, conventional T2WIs were processed with two IHTs (standardization [std] and histogram-matching [HM]) resulting in four paired input image types: initial T2WI, T2WIstd , T2WIHM , and T2-map. VOIs were propagated to extract 45 RFs from MRI-1 and MRI-2 of each image type (LIFEx, v5.10). STATISTICAL TESTS Influence of the input data type on RF values was evaluated with analysis of variance. RFs test-retest repeatability and reproducibility over multiple segmentations were evaluated with intra-class correlation coefficient (ICC). Correlations between k-means clusters and the six tissues depending on the RFs dataset were investigated with adjusted-Rand-index (ARI). RESULTS About 41 of 45 (91.1%) RFs were significantly influenced by the input image type (P values < 0.05), which was the most influential factor on repeatability of RFs (P-value < 0.05). Repeatability ICCs from T2-map displayed intermediate values between the initial T2WI (range: 0.407-0.736) and the T2WIHM (range: 0.724-0.817). The number of RFs with interobserver and intraobserver reproducibility ICCs ≥ 0.90 was 37/45 (82.2%) for T2WIHM , 33/45 (73.3%) for T2WIstd , 31/45 (68.9%) for T2 map, and 25/45 (55.6%) for the initial T2WI. T2 map provided the best tissue discrimination (ARI = 0.414 vs. 0.157 with T2WIHM ). DATA CONCLUSION T2 mapping provided RFs with moderate to substantial repeatability and reproducibility ICCs, along with the most preserved discriminative information. LEVEL OF EVIDENCE 1 TECHNICAL EFFICACY: 1.
Collapse
Affiliation(s)
- Amandine Crombé
- Department of Oncologic Imaging, Institut Bergonié, Comprehensive Cancer Center of Nouvelle-Aquitaine, Bordeaux, France.,Bordeaux University, Bordeaux, France.,Modelisation in Oncology (MOnc) Team, INRIA Bordeaux-Sud-Ouest, CNRS UMR 5251, Talence, France
| | - Xavier Buy
- Department of Oncologic Imaging, Institut Bergonié, Comprehensive Cancer Center of Nouvelle-Aquitaine, Bordeaux, France
| | - Fei Han
- Siemens Medical Solutions USA, Los Angeles, California, USA
| | | | - Michèle Kind
- Department of Oncologic Imaging, Institut Bergonié, Comprehensive Cancer Center of Nouvelle-Aquitaine, Bordeaux, France
| |
Collapse
|
44
|
Samiei S, Granzier RWY, Ibrahim A, Primakov S, Lobbes MBI, Beets-Tan RGH, van Nijnatten TJA, Engelen SME, Woodruff HC, Smidt ML. Dedicated Axillary MRI-Based Radiomics Analysis for the Prediction of Axillary Lymph Node Metastasis in Breast Cancer. Cancers (Basel) 2021; 13:757. [PMID: 33673071 PMCID: PMC7917661 DOI: 10.3390/cancers13040757] [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: 12/16/2020] [Revised: 02/03/2021] [Accepted: 02/08/2021] [Indexed: 12/23/2022] Open
Abstract
Radiomics features may contribute to increased diagnostic performance of MRI in the prediction of axillary lymph node metastasis. The objective of the study was to predict preoperative axillary lymph node metastasis in breast cancer using clinical models and radiomics models based on T2-weighted (T2W) dedicated axillary MRI features with node-by-node analysis. From August 2012 until October 2014, all women who had undergone dedicated axillary 3.0T T2W MRI, followed by axillary surgery, were retrospectively identified, and available clinical data were collected. All axillary lymph nodes were manually delineated on the T2W MR images, and quantitative radiomics features were extracted from the delineated regions. Data were partitioned patient-wise to train 100 models using different splits for the training and validation cohorts to account for multiple lymph nodes per patient and class imbalance. Features were selected in the training cohorts using recursive feature elimination with repeated 5-fold cross-validation, followed by the development of random forest models. The performance of the models was assessed using the area under the curve (AUC). A total of 75 women (median age, 61 years; interquartile range, 51-68 years) with 511 axillary lymph nodes were included. On final pathology, 36 (7%) of the lymph nodes had metastasis. A total of 105 original radiomics features were extracted from the T2W MR images. Each cohort split resulted in a different number of lymph nodes in the training cohorts and a different set of selected features. Performance of the 100 clinical and radiomics models showed a wide range of AUC values between 0.41-0.74 and 0.48-0.89 in the training cohorts, respectively, and between 0.30-0.98 and 0.37-0.99 in the validation cohorts, respectively. With these results, it was not possible to obtain a final prediction model. Clinical characteristics and dedicated axillary MRI-based radiomics with node-by-node analysis did not contribute to the prediction of axillary lymph node metastasis in breast cancer based on data where variations in acquisition and reconstruction parameters were not addressed.
Collapse
Affiliation(s)
- Sanaz Samiei
- Department of Surgery, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (S.S.); (S.M.E.E.); (M.L.S.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (A.I.); (S.P.); (M.B.I.L.); (T.J.A.v.N.); (H.C.W.)
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands;
| | - Renée W. Y. Granzier
- Department of Surgery, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (S.S.); (S.M.E.E.); (M.L.S.)
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands;
| | - Abdalla Ibrahim
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (A.I.); (S.P.); (M.B.I.L.); (T.J.A.v.N.); (H.C.W.)
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands;
- The D-Lab, Department of Precision Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
- Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, Hospital Center Universitaire de Liege, Rue de Gaillarmont 600, 4030 Liege, Belgium
- Department of Nuclear Medicine and Comprehensive Diagnostic Center Aachen (CDCA), University Hospital RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Sergey Primakov
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (A.I.); (S.P.); (M.B.I.L.); (T.J.A.v.N.); (H.C.W.)
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands;
- The D-Lab, Department of Precision Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Marc B. I. Lobbes
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (A.I.); (S.P.); (M.B.I.L.); (T.J.A.v.N.); (H.C.W.)
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands;
- Department of Medical Imaging, Zuyderland Medical Center, P.O. Box 5500, 6130 MB Sittard-Geleen, The Netherlands
| | - Regina G. H. Beets-Tan
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands;
- Department of Radiology, The Netherlands Cancer Institute, P.O. Box 90203, 1006 BE Amsterdam, The Netherlands
| | - Thiemo J. A. van Nijnatten
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (A.I.); (S.P.); (M.B.I.L.); (T.J.A.v.N.); (H.C.W.)
| | - Sanne M. E. Engelen
- Department of Surgery, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (S.S.); (S.M.E.E.); (M.L.S.)
| | - Henry C. Woodruff
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (A.I.); (S.P.); (M.B.I.L.); (T.J.A.v.N.); (H.C.W.)
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands;
- The D-Lab, Department of Precision Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Marjolein L. Smidt
- Department of Surgery, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (S.S.); (S.M.E.E.); (M.L.S.)
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands;
| |
Collapse
|
45
|
Pfaehler E, Mesotten L, Zhovannik I, Pieplenbosch S, Thomeer M, Vanhove K, Adriaensens P, Boellaard R. Plausibility and redundancy analysis to select FDG-PET textural features in non-small cell lung cancer. Med Phys 2021; 48:1226-1238. [PMID: 33368399 PMCID: PMC7985880 DOI: 10.1002/mp.14684] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 01/06/2023] Open
Abstract
Background Radiomics refers to the extraction of a large number of image biomarker describing the tumor phenotype displayed in a medical image. Extracted from positron emission tomography (PET) images, radiomics showed diagnostic and prognostic value for several cancer types. However, a large number of radiomic features are nonreproducible or highly correlated with conventional PET metrics. Moreover, radiomic features used in the clinic should yield relevant information about tumor texture. In this study, we propose a framework to identify technical and clinical meaningful features and exemplify our results using a PET non‐small cell lung cancer (NSCLC) dataset. Materials and methods The proposed selection procedure consists of several steps. A priori, we only include features that were found to be reproducible in a multicenter setting. Next, we apply a voxel randomization step to identify features that reflect actual textural information, that is, that yield in 90% of the patient scans a value significantly different from random texture. Finally, the remaining features were correlated with standard PET metrics to further remove redundancy with common PET metrics. The selection procedure was performed for different volume ranges, that is, excluding lesions with smaller volumes in order to assess the effect of tumor size on the results. To exemplify our procedure, the selected features were used to predict 1‐yr survival in a dataset of 150 NSCLC patients. A predictive model was built using volume as predictive factor for smaller, and one of the selected features as predictive factor for bigger lesions. The prediction accuracy of the both models were compared with the prediction accuracy of volume. Results The number of selected features depended on the lesion size included in the analysis. When including the whole dataset, from 19 features reflecting actual texture only two were found to be not strongly correlated with conventional PET metrics. When excluding lesions smaller than 11.49 and 33.10 mL (25 and 50 percentile of the dataset), four out of 27 features and 13 out of 29 features remained after eliminating features highly correlated with standard PET metrics. When excluding lesions smaller than 103.9 mL (75 percentile), 33 out of 53 features remained. For larger lesions, some of these features outperformed volume in terms of classification accuracy (increase of 4–10%). The combination of using volume as predictor for smaller and one of the selected features for larger lesions also improved the accuracy when compared with volume only (increase from 72% to 76%). Conclusion When performing radiomic analysis for smaller lesions, it should be first carefully investigated if a textural feature reflects actual heterogeneity information. Next, verification of the absence of correlation with all conventional PET metrics is essential in order to assess the additional value of radiomic features. Radiomic analysis with lesions larger than 11.4 mL might give additional information to conventional metrics while at the same time reflecting actual tumor texture. Using a combination of volume and one of the selected features for prediction yields promise to increase accuracy and reliability of a radiomic model.
Collapse
Affiliation(s)
- Elisabeth Pfaehler
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Liesbet Mesotten
- Faculty of Medicine and Life Sciences, Hasselt University, Agoralaan building D, Diepenbeek, B-3590, Belgium.,Department of Nuclear Medicine, Ziekenhuis Oost Limburg, Schiepse Bos 6, Genk, B-3600, Belgium
| | - Ivan Zhovannik
- Department of Radiation Oncology, Radboudumc, Nijmegen, The Netherlands.,Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht, The Netherlands
| | - Simone Pieplenbosch
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Michiel Thomeer
- Faculty of Medicine and Life Sciences, Hasselt University, Agoralaan building D, Diepenbeek, B-3590, Belgium.,Department of Nuclear Medicine, Ziekenhuis Oost Limburg, Schiepse Bos 6, Genk, B-3600, Belgium
| | - Karolien Vanhove
- Faculty of Medicine and Life Sciences, Hasselt University, Agoralaan building D, Diepenbeek, B-3590, Belgium.,Department of Respiratory Medicine, Ziekenhuis Oost Limburg, Schiepse Bos 6, Genk, B-3600, Belgium
| | - Peter Adriaensens
- Hasselt University, Institute for Materials Research (IMO) - Division Chemistry, Agoralaan Building D, Diepenbeek, B 3590, Belgium
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Nazari M, Shiri I, Zaidi H. Radiomics-based machine learning model to predict risk of death within 5-years in clear cell renal cell carcinoma patients. Comput Biol Med 2020; 129:104135. [PMID: 33254045 DOI: 10.1016/j.compbiomed.2020.104135] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/21/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE The aim of this study was to develop radiomics-based machine learning models based on extracted radiomic features and clinical information to predict the risk of death within 5 years for prognosis of clear cell renal cell carcinoma (ccRCC) patients. METHODS According to image quality and clinical data availability, we eventually selected 70 ccRCC patients that underwent CT scans. Manual volume-of-interest (VOI) segmentation of each image was performed by an experienced radiologist using the 3D slicer software package. Prior to feature extraction, image pre-processing was performed on CT images to extract different image features, including wavelet, Laplacian of Gaussian, and resampling of the intensity values to 32, 64 and 128 bin levels. Overall, 2544 3D radiomics features were extracted from each VOI for each patient. Minimum Redundancy Maximum Relevance (MRMR) algorithm was used as feature selector. Four classification algorithms were used, including Generalized Linear Model (GLM), Support Vector Machine (SVM), K-nearest Neighbor (KNN) and XGBoost. We used the Bootstrap resampling method to create validation sets. Area under the receiver operating characteristic (ROC) curve (AUROC), accuracy, sensitivity, and specificity were used to assess the performance of the classification models. RESULTS The best single performance among 8 different models was achieved by the XGBoost model using a combination of radiomic features and clinical information (AUROC, accuracy, sensitivity, and specificity with 95% confidence interval were 0.95-0.98, 0.93-0.98, 0.93-0.96 and ~1.0, respectively). CONCLUSIONS We developed a robust radiomics-based classifier that is capable of accurately predicting overall survival of RCC patients for prognosis of ccRCC patients. This signature may help identifying high-risk patients who require additional treatment and follow up regimens.
Collapse
Affiliation(s)
- Mostafa Nazari
- Department of Biomedical Engineering and Medical Physics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva 4, Switzerland
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva 4, Switzerland; Geneva University Neurocenter, Geneva University, Geneva, Switzerland; Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands; Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
47
|
Abstract
Radiomics describes the extraction of multiple features from medical images, including molecular imaging modalities, that with bioinformatic approaches, provide additional clinically relevant information that may be invisible to the human eye. This information may complement standard radiological interpretation with data that may better characterize a disease or that may provide predictive or prognostic information. Progressing from predefined image features, often describing heterogeneity of voxel intensities within a volume of interest, there is increasing use of machine learning to classify disease characteristics and deep learning methods based on artificial neural networks that can learn features without a priori definition and without the need for preprocessing of images. There have been advances in standardization and harmonization of methods to a level that should support multicenter studies. However, in this relatively early phase of research in the field, there are limited aspects that have been adopted into routine practice. Most of the reports in the molecular imaging field describe radiomic approaches in cancer using 18F-fluorodeoxyglucose positron emission tomography (18F-FDG-PET). In this review, we will describe radiomics in molecular imaging and summarize the pertinent literature in lung cancer where reports are most prevalent and mature.
Collapse
Affiliation(s)
- Gary J R Cook
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK; King's College London & Guy's and St Thomas' PET Centre, St Thomas' Hospital, London, UK.
| | - Vicky Goh
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK; Radiology Department, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| |
Collapse
|
48
|
A FDG-PET radiomics signature detects esophageal squamous cell carcinoma patients who do not benefit from chemoradiation. Sci Rep 2020; 10:17671. [PMID: 33077841 PMCID: PMC7573602 DOI: 10.1038/s41598-020-74701-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 10/06/2020] [Indexed: 11/21/2022] Open
Abstract
Detection of patients with esophageal squamous cell carcinoma (ESCC) who do not benefit from standard chemoradiation (CRT) is an important medical need. Radiomics using 18-fluorodeoxyglucose (FDG) positron emission tomography (PET) is a promising approach. In this retrospective study of 184 patients with locally advanced ESCC. 152 patients from one center were grouped into a training cohort (n = 100) and an internal validation cohort (n = 52). External validation was performed with 32 patients treated at a second center. Primary endpoint was disease-free survival (DFS), secondary endpoints were overall survival (OS) and local control (LC). FDG-PET radiomics features were selected by Lasso-Cox regression analyses and a separate radiomics signature was calculated for each endpoint. In the training cohort radiomics signatures containing up to four PET derived features were able to identify non-responders in regard of all endpoints (DFS p < 0.001, LC p = 0.003, OS p = 0.001). After successful internal validation of the cutoff values generated by the training cohort for DFS (p = 0.025) and OS (p = 0.002), external validation using these cutoffs was successful for DFS (p = 0.002) but not for the other investigated endpoints. These results suggest that pre-treatment FDG-PET features may be useful to detect patients who do not respond to CRT and could benefit from alternative treatment.
Collapse
|
49
|
Shiyam Sundar LK, Muzik O, Buvat I, Bidaut L, Beyer T. Potentials and caveats of AI in hybrid imaging. Methods 2020; 188:4-19. [PMID: 33068741 DOI: 10.1016/j.ymeth.2020.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/18/2022] Open
Abstract
State-of-the-art patient management frequently mandates the investigation of both anatomy and physiology of the patients. Hybrid imaging modalities such as the PET/MRI, PET/CT and SPECT/CT have the ability to provide both structural and functional information of the investigated tissues in a single examination. With the introduction of such advanced hardware fusion, new problems arise such as the exceedingly large amount of multi-modality data that requires novel approaches of how to extract a maximum of clinical information from large sets of multi-dimensional imaging data. Artificial intelligence (AI) has emerged as one of the leading technologies that has shown promise in facilitating highly integrative analysis of multi-parametric data. Specifically, the usefulness of AI algorithms in the medical imaging field has been heavily investigated in the realms of (1) image acquisition and reconstruction, (2) post-processing and (3) data mining and modelling. Here, we aim to provide an overview of the challenges encountered in hybrid imaging and discuss how AI algorithms can facilitate potential solutions. In addition, we highlight the pitfalls and challenges in using advanced AI algorithms in the context of hybrid imaging and provide suggestions for building robust AI solutions that enable reproducible and transparent research.
Collapse
Affiliation(s)
- Lalith Kumar Shiyam Sundar
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | | | - Irène Buvat
- Laboratoire d'Imagerie Translationnelle en Oncologie, Inserm, Institut Curie, Orsay, France
| | - Luc Bidaut
- College of Science, University of Lincoln, Lincoln, UK
| | - Thomas Beyer
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
50
|
Noortman WA, Vriens D, Slump CH, Bussink J, Meijer TWH, de Geus-Oei LF, van Velden FHP. Adding the temporal domain to PET radiomic features. PLoS One 2020; 15:e0239438. [PMID: 32966313 PMCID: PMC7510999 DOI: 10.1371/journal.pone.0239438] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 09/05/2020] [Indexed: 01/18/2023] Open
Abstract
Background Radiomic features, extracted from positron emission tomography, aim to characterize tumour biology based on tracer intensity, tumour geometry and/or tracer uptake heterogeneity. Currently, radiomic features are derived from static images. However, temporal changes in tracer uptake might reveal new aspects of tumour biology. This study aims to explore additional information of these novel dynamic radiomic features compared to those derived from static or metabolic rate images. Methods Thirty-five patients with non-small cell lung carcinoma underwent dynamic [18F]FDG PET/CT scans. Spatial intensity, shape and texture radiomic features were derived from volumes of interest delineated on static PET and parametric metabolic rate PET. Dynamic grey level cooccurrence matrix (GLCM) and grey level run length matrix (GLRLM) features, assessing the temporal domain unidirectionally, were calculated on eight and sixteen time frames of equal length. Spearman’s rank correlations of parametric and dynamic features with static features were calculated to identify features with potential additional information. Survival analysis was performed for the non-redundant temporal features and a selection of static features using Kaplan-Meier analysis. Results Three out of 90 parametric features showed moderate correlations with corresponding static features (ρ≥0.61), all other features showed high correlations (ρ>0.7). Dynamic features are robust independent of frame duration. Five out of 22 dynamic GLCM features showed a negligible to moderate correlation with any static feature, suggesting additional information. All sixteen dynamic GLRLM features showed high correlations with static features, implying redundancy. Log-rank analyses of Kaplan-Meier survival curves for all features dichotomised at the median were insignificant. Conclusion This study suggests that, compared to static features, some dynamic GLCM radiomic features show different information, whereas parametric features provide minimal additional information. Future studies should be conducted in larger populations to assess whether there is a clinical benefit of radiomics using the temporal domain over traditional radiomics.
Collapse
Affiliation(s)
- Wyanne A. Noortman
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, The Netherlands
- * E-mail:
| | - Dennis Vriens
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelis H. Slump
- Robotics and Mechatronics, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tineke W. H. Meijer
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, The Netherlands
| | | |
Collapse
|